You are currently viewing a beta version of our website. If you spot anything unusual, kindly let us know.

Preprint
Review

Review of the Brain Behaviour after Injury and Disease for Its Application in an Agent Based Model (ABM)

Altmetrics

Downloads

197

Views

100

Comments

0

Submitted:

05 December 2023

Posted:

07 December 2023

You are already at the latest version

Alerts
Abstract
The brain being arguably the most complex organ in the human body, its detailed functioning is yet to be fully deciphered, let alone accurately modeled. Nonetheless, the medical community has gathered quite a remarkable amount of studies about the consequences of illnesses and injuries on its development and functioning. This bibliographic review aims to cover mostly the findings related to the changes in the physical distribution of neurons and their connections - the connectome -, both structural and functional, as well as their modelling. It does not intend to offer an extensive medical description of all injuries and diseases affecting the brain, rather presenting the most common ones succinctly so the need for accurate brain modelling can be fully understood and pondered, offering propositions to this aim.
Keywords: 
Subject: Engineering  -   Bioengineering

1. Introduction

The brain acts as an animal’s central computer: it gathers all information obtained through the senses (as a result of external stimuli) and processes it to generate knowledge (through memory) and act upon its surroundings, on top of regulating physiological behaviour and constants like breathing, body temperature or heartbeat; or more vaguely-defined aspects like emotions and conscience. It sends and receives signals through the spinal cord, forming the Central Nervous System (CNS). It is the most energy-consuming organ in relation to its size (a fifth of the total in a resting state), despite undergoing evolutionary optimization for ages in different animal species. [1] Its correct functioning is crucial to the point that a person is considered dead if their brain does not work, although their heart and vital organs could be kept active via artificial means.
According to the World Health Organization (WHO), two of the ten principal causes of death globally in 2019 were directly related to brain malfunction: strokes (also called Cerebrovascular Accidents, CVA) and dementia (including Huntington’s chorea, Parkinson’s, Alzheimer’s and Lewy’s bodies, among many others) [2]. Strokes, ranking second on the list, are caused either by a lack (ischemic, 85% of total [3]) or excess (hemorrhagic) of blood flow in the brain, and thus closely related to the leading cause, Coronary Artery Disease (CAD). Dementia, on the other hand, is an umbrella term for most neuro-degenerative illnesses, resulting in the loss of previously held mental capabilities, especially affecting speech, memory and orientation. They can be age-related (Alzheimer’s, Parkinson’s) or not (e.g.: Huntington’s chorea) and they are not to be mistaken with neurodevelopmental disorders, which affect the brain in its first stages of growth.
Cancer is another prominent cause of death worldwide. Whereas brain tumors are not the most common ones (around one-in-fifty diagnoses [4]), they are amongst the most fatal (one in three five-year survival rate on average in the US, around 5% for the most aggressive types, such as glioblastoma [5]) and alarmingly prevalent in children under 14 (about one sixth of total cases as of 2019, second only to leukaemia [6]). In less lethal but arguably as concerning terms, neurological illnesses varying in cause (including injuries induced by tumors) are the leading cause of crippling disability, and their incidence is increasing, especially affecting ever-aging populations in the developed world [7]. In Psychiatry, the freshly emerged field of Pathoconnectomics explores the links between a defective brain wiring degeneration [8] and disorders like depression [9], schizophrenia or autism [10] or the association between Cerebral Small Vessel Disease (CVSD) and cognitive impairment leading to Alzheimer’s [11].
Neurological disorders are very costly not only in ethical terms but also on an economical level, higher - in absolute terms - dealing in billions of € per year in developed countries where diagnoses are more prevalent [12,13,14,15] but relatively more expensive in underdeveloped countries where means are scarce [7,16,17]. - which poses an inequality issue. Although data itself is relatively abundant, although maybe not representative enough [18,19,20]. In order to manage such valuable and variable data, new approaches have become widespread, such as Data Mining [21,22,23] and Machine Learning [24,25] - especially Graph Neural Networks [26,27,28,29,30,31,32]
Considering all this, it comes as self-evident that the study and characterization of the brain is a great need not only for scientific purposes but also, and most importantly, for health reasons, such as the study and treatment of related injuries and diseases for a sooner and more accurate diagnose and hopefully a more effective treatment. The purpose of this review article is to very succinctly present the most relevant information gathered about such health issues in relation to the brain’s structural and functional architecture. After this introduction underlying the relevance of brain-related illnesses and trauma in Section 1, a brief note on the brain’s structure and function will be introduced in Section 2. Section 3 will present a general overview of the most prominent causes of brain damage, while Section 4 contains some modelling propositions for the connectome. Finally, Section 5 acts as a summary of this article accompanied by some line research suggestions.

2. Mapping the brain: the connectome

The first known reference to the brain is the Edwin Smith Surgical Papyrus (Egypt, 1700 BC), although back then its functions were thought to be carried out by the heart instead, as Aristotle reckoned. Although there were some anatomical studies of the brain during Roman and Baghdad Caliphate times, not much else could be done until the invention of the microscope in the late 1500s AD. Some nerves (especially related to the senses, such as sight or hearing) were correctly identified, but it was not until the second half of the 19th century that the field work by Broca, Wernicke and many others began to correctly associate specific areas of the brain with certain tasks (muscles of speech and language comprehension, respectively).
It became apparent that patients having undergone injury or surgery affecting certain brain zones saw some of their cognitive abilities lessened or fully missing, and so doctors began to fill the blanks in the brain map. One of the first attempts to fully map the brain by function was made by Korbinian Brodmann in 1909 [33], as seen in Figure . Staining neurons with Nissl’s method, he divided the primate brain in 52 areas (44 present in current human beings and 8 remaining only in related primates) based on cytoarchitectural (cell-level structure) criteria.
Brodmann’s map has served as the main guide for decades, and it is still used on an educational level today, but evidence of its incompleteness is abundant and continuously growing. On the one hand, it is possible to live - albeit in an altered state and with great difficulty - with a fraction of a brain, as in severe cases of hydrocephalus - partially substituted by water [34] -, lobotomies - extraction of part of it - [35] or even anencephaly - absence of it altogether - [36]. That can - at least, partially - be explained through the concept of neuroplasticity: the brain’s ability to rewire its functional and structural connections to overcome injury’s effects, hinting that the spatial distribution of functional areas is more flexible than charts like Brodmann’s would suggest. Neuroplasticity allows for a certain regenerative capacity as well, although humbler in the Central Nervous System than in the Periferic one (PNS) [37], which often helps patients recover motor coordination after strokes, for instance [38].
How the brain really works is only partially known to this day, although significant progress has been made since the dawn of Magnetic Resonance Imaging (MRI) in the 1970s, especially its functional variant (fMRI), able to track brain activity in quasi-real time by measuring changes in blood flow [39], assessing pre-surgery conditions [40] and parcelling brain functional [41] and structural areas [42,43,44]- even on a city- [45] or nationwide basis [46,47]. Although relationships between specific tasks and areas in the brain do exist (ref), there is plenty of experimental data suggesting that such a strict function-area mapping framework might not be a good-enough approach. Many individual actions require the activation of multiple areas in the brain (like language and perpetual decision [48]), and the coordination between them (co-activation [49], newly studied with a plethora of techniques [50]), so no area is univocally nor individually responsible for a given action, but rather correlated to it.
Seemingly related (directly or inversely) tasks may be performed by different areas within a brain region, such is the case of facial recognition and evaluation [51]. Activity patterns for the same task can differ [52] and even change with age [53] or illness. Although there’s a certain consensus on a "default" network configuration in resting states [54,55], it is still subjected to individual variations, some of them associated to illness [56]. One must also bear in mind that structural and functional pathways in the brain influence each other. On top of that, most brain mappings show correlation between task and activations, rather than causation of such patterns [57].
Thanks to the experimental work of Santiago Ramón y Cajal in the 1880s, using Nissl’s method, the neuron doctrine gained recognition to become the widely accepted theory that it is today. It conceives the brain (and the whole CNS by extension) as a network of intertwined but independent neurons, a specific type of cell receiving and transmitting electric signals. Neurons are mainly composed of a nucleus (soma), a thin extension covered by myelin (axon) ending in appendices (dendrites) responsible for communication between them (synapse). This connectivity between neurons can be structural - physically existing links between neurons - or functional - the links actually used during neural activity -.
These two sides can be mutually affected and/or change due to injury and/or disease, or even with mere biological age. Neurons themselves can be classified along these lines: structurally as uni/bi/multipolar/anaxonic, etc. or functionally as afferent (sensory), efferent (motor) or interneurons (transition). Furthermore, these connections are subjected to neuroplasticity: rewiring (structural [58]) and alternative paths (functional [59]) life-long [60], especially during and after growth [61], age [62,63], injury [64,65] or disease [66]. The connectome is also affected by diaschisis, by which an area distantly connected to another damaged area might see their functions deteriorated by the latter - a sort of negative functional plasticity [67].

3. Brain Damage

Neural cell death takes place "naturally" in the brain: unneeded neurons are disposed of in a programmed, foreseeable and controlled way - known as apoptosis [68], by which the dying cell implodes collapsing its cytoskeleton and liberating broken nuclear DNA, its remains being eaten by other cells (phagocytosis). This can be a result of brain development [69,70] - well studied and measured [71,72] - and/or due to structural neuroplasticity (axon retraction, i.e. "pruning" [58]) due to redundancy [73]. Apoptosis, be it intrinsic (mitochondrial, BH3-Bax-APAF-1-caspase-9) or extrinsic (death receptor, caspase-8-Bid) [74], is a complex process regulated by multiple internal mechanisms involving various mollecules such as p75, Bcl-2 or caspase-1 [75] and crucial in fighting cancer - which is, at the end of the day, an out-of-control cell multiplication [76,77,78].
Brain damage comes into picture when the unforeseen deterioration or destruction - necrotic death - of brain cells (neurons) takes place. By this process, the cell’s mitochondria and endoplasmic reticulum swell and break the external membrane, due to a plethora of different mechanisms (necroptosis, parthanatos, ferroptosis, pyroptosis, oncosis, lysosomal, autophagic, phagocytic, MitoPore - mitochondrial permeability transition) [74], thus affecting neighbouring neurons though inflammatory expansion - unlike apoptosis. Some of these processes (parthanatos, oncosis, MitoPore) result in ATP (Adenosin-TriPhosphate) depletion - which in turn produce the failure of sodium and calcium channels through the neuron membrane, and with them come cell swelling and degradation via proteases and phospholipases, respectfully. Be it this way or by Mixed Lineage Kinase domain-Like (MLKL: necroptosis), Reactive Oxygen Species (iron: ferroptosis) or inflammation (pyroptosis, lysosomal), the result is abnormal neuron necrosis [74]. As it will be explained in the following subsections, these necrotic mechanisms are linked to the advent of various neurodegenerative diseases [79,80] and other neurological disorders such as sleep apnea [81,82].

3.1. Acquired Brain Injury

Acquired Brain Injury (ABI) is any brain injury caused after birth, and so excluding birth-related trauma and all hereditary or congenital diseases. This exclusion obeys to the fact that the connectome takes years to fully form, so the study of early or pre-natal damage is rather case-dependent, no general procedures having been established. This damage can have an internal (e.g.: tumor growth, mental disorder) or external origin (e.g.: concussion, accident). The first kind are usually - but not exclusively - caused by injuries and are known as to Traumatic Brain Injuries (TBI), while the latter, Non-Traumatic Brain Injuries (NTBI), normally involve a brain illness - not necessarily neurodegenerative. Both have in common widespread destruction of the cortical ribbon and white matter tracts and deep brain damage (thalamus, basal gmanglia) [83]. Other complications incluse hydrocephalus, pneumo-encephalus, ventricular enlargement, skull fracture, unconsciousness, sores, bladder infections, pneumonia and/or multiple organic faliure [83].
Their symptoms can be physical (paralysis, headaches, seizures, insomnia, loss of consciousness, aphasia - speech impediment), cognitive (memory loss, impaired information processing, comprehension or expression), perceptual (disorientation, lack of equilibrium, sight/hearing/touching/smell/taste disfunction, hyperalgesia - extreme pain sensitivity) or behavioural (irritability, aggresiveness, lethargy/apathy).Neural damage, on the other hand, is more difficult to quantify, as most imaging techniques can only identify and count dead neurons [84]. Nonetheless, some techniques do exist [85]. ABI can be primary -shear/tear of tissue, complete right after the impact - or secondary -more complex chemical, biological or biomechanical changes hours after the insult, including blood barrier damage, excitotoxicity (glutamate release), mitochondrial disfunction, Na+/Ca+ influx- [83]. In the most severe scenarios (GCS < 8), it can lead to prolonged coma states or even death.

3.1.1. External: Traumatic Brain Injury

This category includes brain damage caused by physical trauma, i.e. accelerations, shocks, concussions, incisions, etc. caused by a foreign agent. It is responsible for more than 1-in-3 annual injury related deaths in Europe - accounting for 2 million yearly hospitalizations [86] and it affects around 70 million people globally [87] (around 1% of the world’s population), typically young males [88]. TBI can be classified into mild (13-15 points), moderate (9-12 points) or severe (3-8 points) via the Glasgow Coma Scale (GCS), composed of three areas (4 verbal points, 5 ocular and 6 motor). Depending on its causes, it can be further divided into closed (unbroken skull: fast movements, shaking) and penetrating brain injury (open head, e.g. bullets). There are concluding signs of comorbidity with several mental conditions such as Major Depression (MD), Post-Traumatic Stress-Disorder (PTSD), general anxiety and suicidal behaviour [89] - and even sleep disorders, back pain, high cholesterol, osteoarthritis and diabetes [90].
Closed Head Injury
The most common type is mild TBI - meaning not immediately life-threatening (around 80% of all diagnoses [86]), a sub-type of closed head injury (CHI) - occurring within an intact skull - caused by bumps to the head or any other action causing fast skull movement (acceleration/deceleration, especially rotational [91]) which may result in physical strain to the brain and/or chemical changes. These accelerations usually involve coup - contrecoup: back-and-forth jarring of the brain against the skull’s inner walls shearing tissue and resulting in blood vessel rupture, bruising and swelling.
It is relatively common in any physically intensive activity (sports, military practice), acts of violence or accidents (traffic, falls), and can cause some of the aforementioned physical or behavioural symptoms, normally immediately after injury (peaking within the first 24h, during which the brain undergoes gliosis - scar-like multiplication and/or growth of glial cells [91], hindering healing) but entailing potential long-term effects [92,93,94] such as Chronic Traumatic Encephalopathy (CTE) in the case of recurrent milt TBI, typical in sportsmen (up to 90% of athletes in the US [95]) who may experience memory and attention impairments, suicidal behaviour or even cardiovascular complications [96,97].
TBI usually produces three major effects: acute subdural hematoma -associated with traumatic cerebrovascular injury (TCVI) in a limited number of cases (less than 2% [97]), brain contusion and diffuse axonal injury [98]. Contusions involve brain bruising as a result of coup - contrecoup accelerations, mostly affecting the basi-frontal and anterior temporal lobes [99]. Not to be mixed with concussions, an umbrella term for closed-head, mild TBI involving transient mental effects ranging from confusion to loss of consciousness.
Hematomas are extended contusions causing blood overflow from multiple broken blood vessels (due to brain bruising), common in physical trauma. It can be epidural (EDH) or subdural (SDH). The former involves a rapidly leeking broken artery - middle meningeal - between the dura mater (third and most external layer in the meninges) and the skull; whereas the latter takes place when a bridging vein ruptures and slowly seeps between the arachnoid (second meningeal layer) and the dura mater. Traumatic Subarachnoid Hemorrhage (SAH) can also take place if capillaries break and flood the region under that layer. Such blood volumes (be it arterial or venous) may interfere with the Monro-Kellie principle (the total intercranial volume remains constant) [100,101], meaning pressure on the rest of intercranial components, namely brain tissue cerebrospinal fluid (CSF). That could lead to cerebral edema (fluid accumulation), disruption of the blood-brain barrier and/or diffuse axonal injury, among other undesirable effects.
Diffuse axonal injury (DAI) is a the strain/tearing of axons all across the brain due to the stress combination (compression, tension, shear) occurring during and after TBI in 1-15 mm stretches within a particular distribution [83]. It is provoked by both strain (10-50%) and strain rates (10-50 Hz) [98]. This phenomenon is more acute at the junction of gray and white matters with different densities, where the axons are covered in thicker myelin sheaths and surrounded by a drier environment. While and after undergoing stretching, the axon could swell, detach, fracture, increase its permeability and calcium influx and even die (necrosis). [99]. It can also have long-term effects such as a greater chance of developing neurodegenerative diseases such as Alzheimer’s [102,103,104,105]. DAI is usually detected by MRI, although it takes time, so CT may be preferred for fast hemorrhage identification if the patient needs urgent treatment [106], despite CT being less detailed than MRI [83,107], which can be further enriched by Gradient Echo (GRE) [108] or Susceptibility-Weighted Imaging (SWI) [109]. A major problem when modelling DAI is the fact that it is unlocalized, all over the brain (hence "diffuse"), and thus difficult to predict an injury pattern in a deterministic way given a certain traumatic origin.
Penetrating Brain Injury
Penetrating Brain Injuries (PBI), although far less common than Closed Head Injuries (CHI), are also fairly more lethal. They are caused by an external collisions against the skull, which is often fractured - resulting in hematoma and/or intercranial hemorrhage, mostly fatal in the basilar area [88]. Its origins are various (assault, murder/suicide attempts), usually involving physical violence and/or projectiles such as bullets and its consequences harsher: short-term outcomes such as severe trauma in 55% of cases [110] (GCS < 5 for gunshots [111]), generalized hemorrhage, CSF leaking, intracranial infection and aneurysm (50 % lethal [112]) and often death (around 40% [110] - up to 90% if neurological status is poor [112]); and long-term effects like post-traumatic epilepsy and/or seizures for 1 in 2 cases [113].
PBI’s effects vary greatly according to the kinetic energy liberated by the weapon used to inflict the damage E = m v 2 / 2 , which varies linearly with mass (the heavier the object, the greater the damage) and quadratically with its speed (low if under 300 m/s, medium up to 600 m/s and high upwards). Thus, light, low velocity objects like nails or knives are less likely to cause severe damage, and such damage is localized closely to their trajectory. Bullets and shrapnel, on the other hand, travel much faster and can increase their already devastating effect - direct by penetration or indirect by shockwaves (causing cavitation) - depending on shape, angle, deformation or shredding of the skull and/or projectile inside the cranium.
After PBI, rapid evaluation is vital in increasing the chances of patient survival, undergoing CT to determine if surgical intervention is viable, be it to heal wounds and/or extract the projectile (except knifes at first [114]), and act ipso facto to prevent worsening scenarios related to hypoxia, anemia, CSF leaks, hypotension (systolic under 90 mm Hg) or hyperpyrexia (extreme fever over 41,5ºC). Prophylactic anticonvulsants are used to prevent seizures and special attention is paid to aggravating factors: old age, severe coma (GCS = 3), high intracranial pressure, coagulopathy, thrombosis [115]. The prognosis depends greatly on the foreign object’s trajectory, being mostly reserved if it crosses the midline, ventricles or posterior fossa [114] or if it affects the brainstem, both lobes and/or hemispheres [112]. Self-inflicted wounds and pre-hospital intubation and craniotomy/craniectomy are positively correlated with the mortality rate in PBI [110]. Laceration and crushing of tissue as a result of bumping or projectile disintegration enhance damage.
As a result of PBI, neural tissue can be either physically broken (sectioned by the foreign object), impaired or dead (insufficient blood flow due to leakage, hypotension, etc.). Damage is usually more localized than in CHI but also more severe, often meaning lasting neurological consequences and perhaps death. Infections by lack of prophylaxis are another common complication, better explained in the next section as they are largely non-traumatic derived.

3.1.2. Internal: Non-Traumatic Brain Injury

This category includes any brain injury with internal origin, especially involving infections (meningitis, encephalitis, etc.), poisoning (radiation, lead), lack of oxygen (aneurysm, stroke, heart attack) or any other event increasing internal cranial pressure (e.g.: tumors). Sometimes, they can be broadly referred to as ABI, being acute (mainly anoxia/hypoxia-induced: stroke, heart attack, drowning) or chronic (migraines). In general terms, they can potentially stretch all across the brain (diffuse injury) since they target the neuron’s structure [83]. It can have similar effects to TBI, including coma states [116].
Infections
Neurological infections, although almost eradicated in Europe and North America, are relatively prevalent in developing countries. They involve contamination of the cerebrospinal fluid (CSF) in the Central Nervous System (CNS) via blood (hematogenic), contiguity to infected organs or bones or neural transmission [117]. Hematogenic contagion is by far the most usual process, caused by pathogenic agents (bacteria, fungi, protozoa or parasites) in the blood stream, mainly arterial through the junction of white and grey matters (parasitic/bacterial, some viruses) with chances to metastasize elsewhere, although neurotropic viruses (such as herpes or measles [118]) get into the CNS through the blood brain barrier (BBB), transcytotic epithelial passage or leucocyte infestation.
Venous infections are rare, disseminating schistosomiasis (trematode worms) and (micro)thrombophlebitis (blood clot induced vein swelling). Transosteal infections originating in adjacent frontal (face, e.g. sinusitis) or temporal areas (such as otitis affecting the petrous bone) can generate intracerebral abscesses or pericerebral collections (extra-/sub-dural empyema). Neural propagation, although unusual, is a vector for viruses (herpes simplex, varicella, rabies) and bacteria (listeria). Lastly, infection can also occur in direct contact with the cranium or vertebrae after a PBI or surgery (such as nosocomial meningitis). About 1 in 5 brain infection cases have unknown origin [117].
Most common infections imply inflammation (meningitis - meninges, encephalitis - brain parenchyma) or other alterations of intercranial pressure such as abscesses (intracerebral pus accumulation), which of course implies shear and compressive stresses on axons. Some of them (pyogenic or tubercular abscesses, Aspergillosis, Whipple disease, mucormycosis, cryptococcosis, blastomycosis, histoplasmosis, toxoplasmosis, cytomegalovirus, etc.) can complicate diagnosis mimicking space-occupying lesions (SOL, i.e. tumors), losing valuable and timely treatment opportunities [119]. In case of sepsis, damage (inflammation, BBB disruption, hypoperfusion) can have long-term effects (cognitive impairment) [120].
Immunosuppressed patients are particularly vulnerable and carry a worse prognosis. Some Sexually Transmitted Diseases (STDs) such as HIV, herpes, candidiasis, syphilis can have deep neurological impact, most commonly meningitis, but also more severe like Progressive Multifocal Leukoencephalopathy (PML). Some other prominent infections are those related to helminthic worms, provoking angiostrongyliasis, gnathostomiasis, (neuro)cystercosis or schistosomiasis, among others [121]. Over 300 parasites can infect human brains causing eosinophilia (abnormally high concentration of eosinophils in blood [122]). Infections affecting other organs can reach the brain (e.g. tubercolisis expansion causes 6% of all meningitis [123]. Animals can be carriers of viral infections causing encephalitis (rabies by dogs and bats, toxoplasmosis by cats).
Autoimmune diseases
Autoimmune diseases are associated with myelin loss and hemorrhage, come from within [124] or outside the brain (such as lupus erythematosus [125]) and can cause very similar effects to those of infection (meningitis, encephalitis, epilepsy, vasculitis) [126]. Like infections, they often result in brain damage via anoxia, shear/compression or chemically-induced necrosis [124].
Toxic/Metabolic
Neurotoxins causing Toxic Encephalopathy (TE) can be classified into metallic compounds (iron [127], lead, mercury, arsenic, aluminum), nonmetallic inorganic (sulphur, bromine-derived), industrial/environmental (nitric oxid, carbon monoxid, aliphatic hydrocarbons [128], cyanide), nerve agents (gases like sarin, liquids like Novichok), drugs and pharmaceutical products (alcohol, tobacco, cannabis, opiates, cocaine, methamphetamine) and biologic origins (mushrooms - A. Muscaria, A. Phalloides -, animals - snakes, spiders, scorpions -, microorganisms - botulism [129], tetanus, diphtheria -) [130]. Its consequences vary greatly, but often include cerebrovascular accidents (infarct, hypoxia/ischemia, hemorrhage, vasogenic cerebral edema), infection, (astro)glial abnormalities, diffuse neural necrosis and dementia in the long term [131].
This toxins can target the soma (especially heavy metals and Toxic Metabolic Encelopathy), axons (degeneration by industrial chemicals), myelin sheath (separation or loss of myelin sheets resulting in abnormal transmission of nerve impulses), astrocytes (abnormal ionization in membrane activation, processing of toxins). Drugs have a great negative impact on neurotransmission - MDMA disturbs serotonin, methamphetamine kills dopamin [132] - and blood flow in the brain - cocain and heroin are linked to hypoxia and ischemia [133] -. Toxic Metabolic Encephalopathy (TME) can acutely alter neurotransmitters as a result of drug-induced hypoglycemia, anemia, sepsis, anoxia and other organ failures (liver, kidney, lungs), syndromes (withdrawal) [134] and nutritional/endocrine deficits [135].
Radiation poisoning, either accidental or planned (e.g.: radiotherapy as a treatment for tumors) exposure to ionizing agents, can have dreadful effects in brain tissue by four main mechanisms: vascular damage, astrocyte extinction, cytokine alterations and stem cell death (mainly in the hippocampus [136], cerebellum and cortex) [137]. Acute symptoms include edema, headache and drowsiness; mid-term memory loss and attention deficit and long-term vascular hyalinization, myelin depletion, inflammation, ischemia, necrosis, functional damage, dementia [136,138] and even tumor progression instead of its remission - a common adverse effect in oncological radiotherapy [139,140], dose-dependent [141] not so easy to identify [142].
Vascular
Although they can occur after trauma (TCVI [97,143]), cerebrovascular injuries can also be originated from inside the skull by obstruction, breakage, and/or defective oxygenation (hypoxia/anoxia) of blood vessels within brain tissue, leading to necrosis: a stroke, the second leading death cause worldwide [2,144]. Also known as Cerebrovascular Accident/Insult (CVA/CVI) or apoplexy, its incidence is steadily increasing (especially for population over 75 years old). It can be ischemic (brain infarction due to insufficient blood supply via vessel occlusion) or hemorrhagic (intracranial blood spilling via vessel or aneurysm rupture).
The former are by far the most common - around 85% of all cases [144,145] - and it is sometimes anticipated by Transient Ischemic Attacks (TIA) [145], falling into several etiological categories: large-artery artherosclerosis, cardiogenic embolism, small vessel occlusive disease, determined or undetermined cause. Hemorrhagic stroke can be further classified into intracerebral (ICH, within brain) and subarachnoid (SAH, between inner and outer meninges), as previously mentioned. Its symptoms include half-body paralysis, high blood pressure, arm stiffness, headache and incoherent speech or motor impairment. Risk factors can be fixed - age, genetics (race, family history), sex - or modifiable - hypertension (causing hemorrhagic strokes), diabetes, nutrition (diet, obesity), alcohol, sedentary lifestyle or cardiac conditions among others - [144,145].
Strokes disturb the brain’s functional connectivity within minutes [146], either by tissue loss (necrosis) or diaschisis, as well as abnormal patterns such as trading interhemispheric connectivity for intrahemispheric among usually unrelated local networks [147]. It is as well a key element for recovery [148,149,150] by promoting small-worldness in neural networks, i.e. high nodal clustering (specialization) and short inter-nodal path lengths (efficiency, integration) [151]. Studying the brain’s angiome (blood vessel network) can provide meaningful information when facing stroke detection and therapeutics, since its mappings are easier and more developed than those of the connectome [146] - which it directly affects.
Cancer
Tumors, also known as neoplasms, are lumps of abnormally - and uncontrollably - growing cells. The can be benign (non-cancerous, slowly growing and unlikely to spread) or malign (cancerous, rapid growth and prone to propagate). Brain tumors originate mainly in intercranial tissue or the meninges (meningiomas), although they mostly [152] come from different adjacent organs (metastasis); such is the case of melanoma (skin cancer), prostate/breast cancer, lung cancer or Hodgkin’s lymphoma (lymphatic system cancer). Although rare amongst cancers (2% of total [4]), brain tumors have a great potential to be lethal (around 10% 10-year survival rate [4]) or produce long-term sequels. Risk factors include ionizing radiation, genetics (hereditary syndromes: neurofibromatosis, tuberous sclerosis), immunosupression (infection, allergy), chemicals (N-nitroso compounds are mutagenic and trespass the BBB) and head trauma.
Symptoms are very generalistic (nausea, headaches, fatigue), although the apparition and evolution cognitive impairment (aphasia, hemiparesis, motor dysfunction) and seizures are usually more telling [153]. Diagnosis is made through gadolinium-contrasted MRI - commonly when the BBB barrier is already broken by a malignant neoplasm, since most radiotracers used for other tumors - which, of course, poses a limitation to act upon the injury. Nonetheless, some alternatives like fluorodeoxyglucose (FDG) and labeled aminoacids might be able to detect low-grade gliomas in time before they develop into glioblastomas [154].
There are more than 100 types of brain tumors, and their classification is difficult, having undergone recent restructuration to include molecular biology - namely, mutations related to proteins IDH, B-raf or MGMT - in their traditionally histologic criteria [155]. If they originate within the CNS, they are called primary brain tumors, the most common being gliomas (75% of diagnoses [156]) originating in glial cells or supportive tissue in a circumscribed or diffuse manner: astrocytomas on astrocytes (anaplastic - grade III, glioblastoma multiforme - grade IV), ependymomas (ventricles, spinal cord), oligodendrogliomas (CNS, myelin production), or brain stem gliomas. Some of them can be localized (astrocytomas), while others are diffuse and can easily extend (glioblastoma multiforme) [153]. Other brain tumors without glial origin are medulloblastomas (developing nerve cells), meningiomas (meninges), schwannomas (Schwann cells), craniopharyngiomas (pituitary, near hypothalamus) and germ cell (gametes) tumors.
Their effects on the connectome are multiple: mechanical (pressure by tumor growth on the remaining healthy brain tissue plus sometimes edema, damaging it), chemical (protein mutation deteriorating synapses), vascular (thromboembolism) and necrosis - with the subsequent functional impairment depending on the area. As mentioned above, neurotoxicity is both a common origin and byproduct of some tumor treatments (radiotherapy, chemotherapy) - identified as a possible trigger for glioblastoma [157].

3.2. Neurological disorders

Diseases are a common origin for brain damage, not injury-related but rather result of a prolonged condition, developing in a mid/long-term basis. They are presented in a separate group to differentiate them from incidental, acute insult (previous section). They include pathologies like epilepsy and dementia, neurodegenerative illnesses and psychiatric/motor disorders.

3.2.1. Epilepsy

Epilepsy is a chronic, non-contagious neurological disorder causing recurrent (at least two within 24 hours) uncontrollable, unprovoked seizures - thus excluding those induced by traumatic brain injury, electrolyte disequilibrium or concomitant diseases. It affects 1% of world population [158] and its onset can be focal, generalized - affecting both hemispheres, motor or non-motor - or unknown. Etiologically, it can be genetic (mutation affects management of voltage-dependent sodium channels [159]), structural (network abnormalities), infectious, metabolic (biochemical changes), immune (induced inflammation) and unknown. Although it is rarely lethal, epilepsy can have long lasting neurological effects [160].
Its incidence depends on age (prominent during childhood - 75% of diagnoses [161] - and adolescence), gender (more common in males), availability of medical devices (compromising in developing countries, more diagnoses and generalized rather than focal seizures [158]), socioeconomic status (the lower the income, the higher the chances) [162] and developmental conditions (autism is correlated). Its symptoms vary greatly with the originating cortex area: visual phenomena - blinking, vision loss, hallucinations - (occipital lobe), clonic/tonic motor responses (precentral gyrus, frontal lobe), sensory - e.g. numbness (postcentral gyrus), etc. [161] They may be preceeded by a warning or omen, known as "aura".
Epileptogenesis involves an imbalance between excitative and inhibitive neuronal pathways as a result of uneven activation potentials creating a synchronized wave of excessive neuron firings [158,159]. Malfunction in enzymes (ATPase) and/or glia regulating the extracellular ion concentration (e.g.: potassium overload) may produce neuron depolarization and so action potential discharge leading to seizures, which in turn raise even higher the potassium levels; creating a cascade effect (epilepsy) - which also strengthens dendritic echoes, further enabling activation [163]. GABA-ergic synaptic transmission may lead to depolarization if its controlling ions (e.g. chloride) are altered. Aberrant neural network synchronization requires not only the aforementioned excessive discharge but also triggering events such as paroxysmal depolarization shifts (PDS) of cortical pyramidal cells or neuroplasticity itself - for instance, axon collateral sprouting [159]. Although anti-epileptic drugs are effective in 2 out of 3 cases [164], insight in the connectivity implications of epilepsy [165] is greatly needed for effective treatment strategies. Synchronization may be limited to the lamellar axis, according to some studies [166].

3.2.2. Neurodegenerative diseases

Neurodegenerative diseases are becoming increasingly common worldwide, especially in developed countries where life expectancy is higher and thus dementia is more likely to appear [167]. They entail a progressive, inevitable deterioration and ultimate death of cells within the nervous system (both central and peripheral, though mostly in the brain [168]) causing neurological dysfunction, ranging from memory loss - e.g. Alzheimer’s - to motor impairment - sclerosis, Parkinson’s -. They can be classified according to etiology into amyloidoses (Alzheimer’s, Creutzfeldt-Jakob’s), tauopathies (Pick’s disease, CTE), alfa-synucleinopathies (Lewy bodies, multiple system atrophy) and TDP-43 proteinopathies (some types of sclerosis) [169].
Although very diverse in nature, they all share some common features. Firstly, instead of a static neuronal loss - typical for metabolic/toxic NTBI -, necrosis progressively affects certain cells due to their vulnerability and it spreads according to the brain’s structural and functional connectivity [169] (neural pathways). Secondly, they are related to a chronic immune malfunction. Lastly, they generally cause inflammation. [170]
Dementia-type Dementia is the gradual loss of mental skills such as memory, thinking, learning and judgement. It affects around 50 million people globally - mainly over 65 years [171] with symptoms such as behavioural changes and/or deficits in communication, orientation and memory. While cell deterioration is part of the normal ageing process, dementia implies There have no definitive cure - the only attainable goal being the slowing of the decay - and ultimately make the patient dependent on others. The main risk factors are lower education, hypertension, hearing impairment, smoking, obesity, depression, sedentary lifestyle, diabetes, isolation, TBI, alcohol and air pollution [172]. Most common illnesses causing dementia are Alzheimer’s and Lewy’s bodies, although its origins can also be vascular (strokes), STDs or trauma (CTE). Dementia is not to be mistaken for delirium, which is sudden, transitory and mainly distorts attention mechanisms.
Alzheimer’s Disease is the most prominent dementia-inducing disease (half of all cases [173]), affecting around 5% of the European population [174] - the most deeply affected continent due to its advanced life expectancy. A- β plaques and neurofibrillary tangles (NFTs) are generated by accumulation of amyloid β -peptides and hyperphosphorilated τ -proteins (and/or demyelinization [175,176]), causing inflammation and degeneration of the brain tissue and so excessive, non-programmed cell death (necrosis) and volume changes (gyri shrink, sulci grow - overall volume loss up to 50%) [80]. It can also be the result of mutation (genes APP, PS1, PS2), though very rarely (less than 1 in 20 cases).
AD deteriorates memory, thinking processes and alters emotions by provoking diffuse neuronal loss, synaptic degeneration (correlated with NFT distribution) and reactive gliosis (abnormal glial and astrocyte growth) [80] altering the complex cellular micro-environment in the brain [177]. It starts as a localized neural loss (locus ceruleus, entorhinal cortex) but eventually becomes diffuse (amygdala, hippocampus, frontal/parietal cortices). As neurons die all over the brain, paths between them get longer while local clustering remains in the unaffected areas, yielding an efficiency loss as "small-world" characteristics fade away [178] - selective hub vulnerability [179] - although that depends on the characterization of such graphs (sample size, brain area, measurements) [180].
The disease’s progress can be tracked by synaptic activity, namely the post-synaptic density protein PSD-95 [181,182,183] and other biomarkers [184]. Enhancing neuroplasticity [66] and some immunotherapeutic drugs [184] or boosting the endogenous cannabioid system (ECBS) in the brain [185,186] can help slow down the advancement of AD, especially in its prodromal (i.e. early) stages. A precocious diagnosis [184] and realistic computational approaches [179] can prove important in treatment.
Dementia with Lewy Bodies (DLB) is the result of the accumulation of the alpha-synuclein protein in the brain, creating deposits (Lewy bodies) which disturb the brain’s chemical balance. Although not as common as AD, it represents a sizeable share of cases (around 5-7,5% of all dementias [187]). It is one of two diseases caused by Lewy Bodies, the other being Parkinson’s (PD), discussed in the next section.
Movement disorders Under a relatively new and phenomenological medical category [188], this section encompasses the most common neurodegenerative diseases affecting motor abilities, i.e. walking, reflexes, standing, etc. as a result of a disruption or dysfunction of coordination between the CNS (brain, spinal cord) and muscles. According to the movement’s disruptive expression, they can be further subdivided into hyperkinesias (excessive), dyskinesias (unnatural) - jerky or non-jerky -, hypokinesias (decreased reach), bradykinesias (slowness), akinesias (absence) and abnormal involuntary movements [188]. Although this subsection focuses on prolonged disorders prominent in advanced ages (+65 years), transitory movement disorders - such as tremors, dystonia or tics - are not uncommon in younger patients and can result in early Parkinson’s misdiagnosis [189].
Parkinson’s Disease (PD) produces hypokinesia and bradykinesia - among other non-motor symptoms - stemming from Lewy’s bodies ( α -synuclein deposits), presenting high comorbidity with LBD and so greatly hampering an accurate (and timely) diagnosis [187] - which already requires very high-fidelity MRI [190]. This deposits provoke localized necrosis in the ventrolateral substantia nigra - namely targeting dopaminergic neurons - and so dopamine underflow to the striatum within the brainstem [191]. About 10-15% of all cases are genetic, and they can originate from prion diseases and perhaps metabolic iron accumulation [192] as well.
PD results in axonopathy, demyelination [193] (particularly originating in the basal ganglia [194]) and synaptic dysfunction and reduced white matter structural connectivity [195] - especially along certain pathways: nigro-pallidal, frontoparietal-stratal, etc. Some studies suggest this connectivity decrease and subsequent cortico-striatal topological reorganization is present in prodromal symptoms of PD, like rapid eye movement sleep behaviour disorder (iRBD) [195]. It also disrupts functional connectivity, although reliability and reproducibility between patients undergoing different stages, treatments and severities of PD and time evolution is problematic when comparing to healthy individuals [196], although some praiseworthy attempts exist [197]. Treatment usually involves L-DOPA (levodopa or l-3,4-dihydroxyphenylalanine) which crosses the BBB to increase dopamine concentrations in the brain.
Lastly, one must not mistake PD for ataxia, which is an acute lack of coordination of different muscles affecting gait, speech and eye movement among others as a result of nervous damage, mainly cerebellar and reversible in some cases [198].
Huntington’s Disease (HD) is a rare, genetic (autosomal dominant) disorder affecting the CNS and showing symptoms like chorea (involuntary, fast and abrupt movements of facial, trunk and/or limb muscles), behavioural and psychiatric degeneration - including dementia -. It can manifest itself at any point in the patient’s life, with no noticeable clinical indices until then [199], although most diagnoses happen between 30 and 50 years of age, inevitably leaving to full dependency and death (most commonly by pneumonia or suicide) [200]. It is caused by a cytosine-adenine-guanine (CAG) trinucleotide repeat (more than 36 times) in the huntingtin (HTT) gene on chromosome 4p and frequently treated with dopamine receptor blockers [200].
In terms of structural connectomics, HD greatly decreases nodal betweenness centrality (i.e., reduced relative importance of certain nodes within the network) and the clustering coefficient - meaning an impaired capacity for inter-nodal information processing [201] that can spread throughout the brain as the mutant protein propagates, shaping propagation patterns - with origin in the striatum and explaining white and grey matter deterioration - predictable via graph theory [202]. HD disrupts as well functional connectivity in subcortical and default mode networks. In some brain areas (within putamen, insula-putamen, visual networks), functional connections are further impeded as the CAG repeat length increases; whereas the contrary is true for others (calcarine to middle frontal gyri) [203].
Prion diseases, also known as transmissible spongiform encelopathies (TSE) are rare neurodegenerative, deadly illnesses caused by misfolded proteins (prions) P r P C mutating irreversibly into P r P S c and causing neuronal necrosis, vacuolation and abnormal activation of microglia and astrocytes [204]. This can take place in humans and other animals (e.g.: sheep, deer, cows), in all organs but mainly the brain and the CNS. Their incubation process is long (up to decades), during which those proteins accumulate and create microscopic holes in the brain, transforming its tissue into a sponge (hence the name).
Prion diseases (PrD) can be divided into sporadic (spontaneous and unpredictable: Creutzfeld-Jakob disease (CJD), fatal insomnia, variable protease sensitive prionopathy), familial (genetically transmitted: familial CJD and fatal insomnia, Gerstmann – Sträussler – Scheinker disease) and acquired (through introduction of contaminated tissue into the patient - ingestion, transfusion, etc.: kuru, variant CJD). CJD is the most common prion disease, affecting 1 in a million yearly (85% sporadic, 10-15% familial [205]), manifesting itself in young adulthood if acquired (vCJD) and senescence if sporadic (sCJD) [206]. Although uncommon and heterogeneous in etiology and diagnosis, the study of prion diseases can potentially shed light on the role of protein misfolding in more widespread neurodegenerative diseases such as Parkinson’s and Alzheimer’s [207] and its transmission along connected structural pathways, which can be modeled as graph diffusion [208].
Multiple sclerosis (MS) can be also considered a neurodegenerative disease [209,210] in its latest stages, after initial autoimmune inflammation in the CNS [170,211,212]. "Sclerosis" means "abnormal hardening of body tissue". Its ultimate etiology is unclear, but it involves multiple genes increasing susceptibility along some environmental (ultraviolet B exposure), disease (Epstein-Barr virus) [212] and genetic factors (highest incidence among European and North American populations [210,211]). It is usually diagnosed in early adulthood via MRI (several white matter scars/plaques over time, chronic CNS inflammation) and it can be intermittent (relapsing-remitting, 85% of cases) or chronic, with chances of drug-induced remission (secondary progressive MS) or not (primary progressive or progressive-relapsing MS) [211].
MS affects mainly optic nerves, the brainstem and the spinal cord [212]. comprise demyelination and neuronal loss through axon deterioration [211] so, although remission can happen within hours or days, it is never complete because the neuronal reserve is progressively depleted - hence the neurodegenerative nature of the illness, despite partial remyelination [212]. Primary progressive MS entails ataxia and progressive cognitive and visual failure [212]. Lesions appear as the illness advances, affecting the connectome’s structure (inflammation, atrophy) and thus also its functioning: abnormal activation of frontal regions or hippocampus for memory tasks and upsetting the default mode network (DMN) in resting states - although its direct links to cognitive impairment have proven difficult to clarify [213,214].
Functional connectivity remains a benchmark for studies about MS nonetheless, with special attention to network efficiency indicators [215,216] - e.g. on working memory, but subjected to patient heterogeneity [217] - with an apparent common ground altogether: altered connectivity in deep-gray matter areas, lower brain modularity, hemispheric skewness and task-independency [218]. Treatment strategies have rapidly improved in a paliative sense but keep failing to put a remedy to continuous neurodegeneration [219].
Amyotrophic lateral sclerosis (ALS) - also known as Lou Gehrig’s disease - is a rare (1 in 100000 [220]) neurodegenerative disease targeting mainly motor neurons in the brain (upper, in the frontal lobe) and the spinal cord (lower) and the brainstem. As such, it is incurable and, on average, patients perish 2 to 5 years after diagnosis (usually after 60 years [220]) due to the malfunction of the diaphragm (breath) and/or swallowing muscles (nutrition) - with notable exceptions of longevity such as Sir Stephen Hawking. Although it starts focally, ALS often spreads to other body parts. It etiology can be autosomal inheritance (10 %) by a hexanucleotide repeat expansion of gene C9orf72 (between 1/3 and 1/2 of familial cases, although there are at least other 25 genes involved [221]) or sporadic (unclear). Half of all patients experience extra-motor conditions like behavioural changes, language impairment or executive dysfunction, and 1 in 10 show signs of frontotemporal dementia [222].
It is proven that brain topology worsens quantitatively (leaf fraction, degree divergence and correlation) in terms of efficiency as ALS unfolds, disentangling the multiscale, assortative networks - bonding between similar nodes - typical for a healthy connectome [223] - which translates into a functional impairment accordingly, predictably with more intensity within the motor cortex [224]. Overall, patients show a decreased functional connectivity in the cortex (right orbitofrontal, left interior frontal) and the corpus callosum [225] as well as an enhancement in the right angular, parietal cortex and the frontoparietal networks [226]. All this changes could be interpreted as compensatory mechanisms - surrogates for lost or damaged brain regions.

3.3. Psychiatric disorders

This category includes non-neurodegenerative disorders affecting mental health, also referred as mental disorders and illnesses and commonly treated by psychiatrists. They can be defined as clinically significant disturbances in an individual’s cognition, behaviour or emotional processes affecting their normal mental function; usually listed on the Diagnostic and Statistical Manual of Mental Disorders (DSM). They are projected to be one of the biggest health concerns during this century and they usually develop from infancy and early adulthood (by mid-20s) [227], while diagnoses peak around 15 years (early adolescence) [228].
An early treatment could very much determine the future evolution of the illness and life quality in the subsequent years. Neurodevelopmental disorders (mental disability) and anxiety are diagnosed in early infancy (around 5 years), while obsessive-compulsive and eating disorders bloom during adolescence (around 15 years) and long-lasting issues like addiction/substance abuse and psychiatric disorders (mood, psychosis/schizophrenia, personality) appear evident in the first 20s [228].
Unlike neurological disorders such as the ones listed in the previous section, they are purely behaviourally diagnosed. Connectomics has shifted the paradigm in psychiatric studies as it allows for the identification of structural and functional measurable changes in the patient’s brain during and after illness on a graph-like basis [229], such as local network efficiency, clustering coefficients and global communication path lengths [230]. MRI has been particularly useful in mapping the cortical surface, structural (diffusion, dMRI) and functional (fMRI) connectivity patterns - even in resting state (rfMRI), although coactivations for the same task and a great individual variability must be considered [231]. Alterations in structural and functional connectivity in the brain have been associated to schizophrenia, major depression, bipolar disorder and autism, among other disorders [232].
Schizophrenia is a chronic mental health disorder affecting 1% of the population and growing in incidence in developed countries [233], whose symptoms can be considered "positive" - hallucinations, voices, paranoid delusions - or negative - social handicap, loss of affection, drive or sense of pleasure, incoherent speech, erratic behaviour and diminished cognitive ability - [234]. It may occasionally relapse (via therapy and drugs) or become incapacitating.
It provokes a neurochemical imbalance by which the normal course of neurotransmitters (dopamine, serotonin, glutamate, perhaps aspartate, glycine or GABA) is altered, mainly along four pathways: nigrostriatal (caudate nucleus to substantia nigra) - linked to motor symptoms -, mesolimbic (ventral tegmental to limbic areas), mesocortical (ventral tegmental area to cortex) - associated with negative symptoms as the dopamine level decreases - and tuberoinfundibular (hypothalamus to pituitary gland) [235].
Its etiology is manifold, from obstetric problems (infections, fetal complications) to genetic factors (parents, monozygotic twins) and environmental triggers (social isolation, trauma). The connectome undergoes important changes during the illness, such as left frontal lobe hyperactivity [236], network randomization as clustering decays due to neural path over-shortening [237] - distortion of the characteristic "small-world" properties in healthy brains [238] and altered hierarchies in functional connectivity [239].
Major Depression (MD), also known as Major Depressive Disorder (MDD), is a persistent (at least 2 weeks) and debilitating sensation - with apparent cause or not - of sadness or melancholy associated with a loss appetite, libido, interests, pleasure sensations (anhedonia) and/or sleep, cognitive impairment and irritability [240]. It affects 1 in 6 people along their lifetimes [241]. It is the second most devastating burden in terms of disability-adjusted life years (DALY), which includes years lived with disability (DLY) - which can last all life - and years of life lost to premature mortality (YLL) - mainly due to suicide (50% of them are depression-induced), but also as a result of comorbidity with diabetes, heart diseases and stroke. It has a large genetic component (about 35% [240,241]) - although not attributed to any specific gene single-handedly - which more often than not comes with an array of reinforcing behavioural traits (conflict avoidance, pessimism, anxiousness) [240].
MDD is responsible for a decrease in functional connectivity among fronto-amygdalar [242], somatomotor, dorsal attention [243,244], salience - relevant for treatment [245], executive control and default mode networks in resting state [246] - linked to social dysfunction [247] and potentially recoverable by electro-convulsive therapy (ECT) [248] and medication [249] - predicting relapse [250]. Despite that, this alternations do not affect the global network irreversibly [251].
In combinations with manic episodes (grandiosity, good mood, overconfidence), MDD produces bipolar disorder (BB) [252], which exacerbates this mood swings turning them into extenuating chronic cycles that may become crippling. According to the severity of said manic events, experts separates bipolar I (acute, severe, followed by delusions and hallucinations in 75% of cases), bipolar II (less severe, hypomania) and cyclothimic disorder (recurring depressive and hypomanic states for over 2 years) [253]. In all three versions, it affects 1 in 100 people worldwide and lists among the leading causes of disability for young patients, yielding cognitive and behavioural impairment that can induce cardiopathies [254] lead to suicide during the worst episodes [255]. Connectivity distortions are similar to those of MDD, adding an important decoupling between functional and structural pathways - strongly correlating with suicide attempts [256,257].
Autism Spectrum Disorder (ASD), formerly known as Pervasive Developmental Disorder (PDD) [258,259] or "childhood schizophrenia" [260], encompasses conditions involving a combination of social communication and recurring behaviours, commonly with an early onset (around 3 years of age) and followed by sensory anomaly and perhaps intellectual disability. It concerns about 1 in 100 people [261] worldwide, although prevalence is higher in rich countries [259] as the definition and diagnose methodology has shifted in the last decades [262]. It is namely hereditary - with a plethora of associated genes and proteins [263], some of them locally inhibiting neural connectivity [264,265] - and presents high comorbidity with other neurological (ADHD, epilepsy) or psychiatric disorders (depression, anxiety) [259].
In contrast to most disorders disclosed in this article, it has been found - through fcMRI, MEG and EEG - that ASD appears to enhance functional connectivity excessively (hyperconnectivity) on a network and state base, depending on age as well - although some life-long effects exist, e.g. parietal and frontal hyperactivity (linked to repetitive behaviour in early development) and long-distance hypoactivity [266],depending on the analyzed frequency [264]. Nonetheless, this hyperactivity is not necessarily reflected on a structural basis - apart from higher gyrification, rather the inverse: brain overgrowth during development makes adults with autism have a delayed long-distance connectivity (less efficient). Despite the general tendencies, individual variability [267] and symptom severity [268] play a crucial role on connectivity. In spite of proof of increased network efficiency markers such as betweenness centrality [269,270], there is contradictory evidence in regards to ASD-induced frontal hyperactivity [271].
Asperger syndrome (AS) is a chronic neurodevelopmental disorder related to, if not included, in the autism spectrum [272], presenting similar symptoms and connectivity disturbances to those of broad "autism", except for a greater intelligence on average and general absence of dysphasia (impaired propositional language) [273], rather the contrary as patients develop quite a structured form of language - albeit delivered in uncommon ways [272]. Differential diagnosis from social phobia and schizoid personality can prove difficult [274]. There is no structural difference in the brain between healthy controls and AS patients, while functionally they have higher global efficiency (greater transfer speed) and lower network segregation (transitivity) and resilience (assortativity) [275].
Obsessive-Compulsive Disorder (OCD) is a mental health condition described by repetitive, ritualistic behaviours involving checking tasks and washing excessively; feeling anxiety and fear if they do not perform in such manner. Appearing in early adulthood (22-36 years), OCD involves intrusive thoughts and is often misdiagnosed. It is not curable and may be genetically inherited, though treatment - normally serotonin re-uptake inhibitors and cognitive–behavioural therapy, perhaps surgery [276] - reduces symptoms greatly improving daily life functionality [277].

4. Conclusions and future research lines

Biophysical models, especially those allowing for brain activity tracking, can greatly enhance diagnoses of neurodegenerative illnesses [168,278].
Final objective: customized modelling and diagnose (individual, real-time brain mapping).
Extend analysis to all Central and Periferic Nervous Systems, considering the role of glia, myelin, angiome, etc. Focus on the cortex (most synapses within 20% of the brain’s volume) and cerebellum (80% of all neurons contained in 10% volume) [231].
Emphasis on preventive monitoring and more accurate diagnoses (Digital/Hybrid Twins [279])

Funding

This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Słodowska-Curie Grant Agreement No. 956401 Grant PID2021-126051OB-C43 funded by MCIN/AEI/ 10.13039/501100011033 and by “ERDF A way of making Europe”.

Acknowledgments

The authors would like to thank Professor Michel Destrade (Chair of Applied Mathematics, University of Galway) and Sairam Pamulaparthi Venkata (Early-Stage Researcher 8, XS-Meta Project, University of Galway) for their insights on brain modeling as a soft, visco-elastic material, as well as Itziar Terradillos Irastorza (PhD in Neuroscience 2021, UPV/EHU), Hanoi Iván Guillermo Montiel (PhD student, Institut Pasteur) and Edgar Soria-Gómez (Neuroscience Department at UPV/EHU) for their medical guidance.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Padamsey, Z.; Rochefort, N.L. Paying the brain’s energy bill. Current Opinion in Neurobiology 2023, 78, 102668. [Google Scholar] [CrossRef] [PubMed]
  2. Ritchie, H.; Spooner, F.; Roser, M. Causes of death. Our World in Data.
  3. Murphy, S.J.; Werring, D.J. Stroke: causes and clinical features. Medicine 2023, 51, 602–607. [Google Scholar] [CrossRef]
  4. McKinney, P.A. Brain tumours: incidence, survival, and aetiology. Journal of Neurology, Neurosurgery and Psychiatry 2004, 75, ii12–ii17. [Google Scholar] [CrossRef] [PubMed]
  5. Miller, K.D.; Ostrom, Q.T.; Kruchko, C.; Patil, N.; Tihan, T.; Cioffi, G.; Fuchs, H.E.; Waite, K.A.; Jemal, A.; Siegel, R.L.; Barnholtz-Sloan, J.S. Brain and other central nervous system tumor statistics, 2021. CA: A Cancer Journal for Clinicians 2021, 71, 381–406. [Google Scholar] [CrossRef] [PubMed]
  6. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. Journal of Epidemiology and Global Health 2019, 9, 217. [Google Scholar] [CrossRef] [PubMed]
  7. Feigin, V.L.; multiple collaborators. Global, regional, and national burden of neurological disorders, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. The Lancet Neurology 2019, 18, 459–480. [Google Scholar] [CrossRef]
  8. Pfeiffer, R.L.; Anderson, J.R.; Dahal, J.; Garcia, J.C.; Yang, J.H.; Sigulinsky, C.L.; Rapp, K.; Emrich, D.P.; Watt, C.B.; Johnstun, H.A.; Houser, A.R.; Marc, R.E.; Jones, B.W. A pathoconnectome of early neurodegeneration: Network changes in retinal degeneration. Experimental Eye Research 2020, 199, 108196. [Google Scholar] [CrossRef]
  9. Liu, W.; Wang, X.; Hamalainen, T.; Cong, F. Exploring Oscillatory Dysconnectivity Networks in Major Depression During Resting State Using Coupled Tensor Decomposition. IEEE Transactions on Biomedical Engineering 2022, 69, 2691–2700. [Google Scholar] [CrossRef]
  10. Rubinov, M.; Bullmore, E. Fledgling pathoconnectomics of psychiatric disorders. Trends in Cognitive Sciences 2013, 17, 641–647. [Google Scholar] [CrossRef]
  11. Dey, A.K.; Stamenova, V.; Turner, G.; Black, S.E.; Levine, B. Pathoconnectomics of cognitive impairment in small vessel disease: A systematic review. Alzheimer’s and Dementia 2016, 12, 831–845. [Google Scholar] [CrossRef]
  12. Olesen, J.; Gustavsson, A.; Svensson, M.; Wittchen, H.U.; Jönsson, B.; and. The economic cost of brain disorders in Europe. European Journal of Neurology 2011, 19, 155–162. [Google Scholar] [CrossRef]
  13. DiLuca, M.; Olesen, J. The Cost of Brain Diseases: A Burden or a Challenge? Neuron 2014, 82, 1205–1208. [Google Scholar] [CrossRef] [PubMed]
  14. Parés-Badell, O.; Barbaglia, G.; Jerinic, P.; Gustavsson, A.; Salvador-Carulla, L.; Alonso, J. Cost of Disorders of the Brain in Spain. PLoS ONE 2014, 9, e105471. [Google Scholar] [CrossRef]
  15. Pino, R.D.; Díez-Cirarda, M.; Ustarroz-Aguirre, I.; Gonzalez-Larragan, S.; Caprino, M.; Busnatu, S.; Gand, K.; Schlieter, H.; Gabilondo, I.; Gómez-Esteban, J.C. Costs and effects of telerehabilitation in neurological and cardiological diseases: A systematic review. Frontiers in Medicine 2022, 9. [Google Scholar] [CrossRef] [PubMed]
  16. Faruqi, S.J.; Shahbaz, N.N.; Nisa, Q.; Umer, S.R.; Ali, S.G.; Aziz, M.Y. Cost of Investigating Neurological Disease: Experience of a Tertiary Care Center in Karachi, Pakistan. Cureus 2020. [Google Scholar] [CrossRef] [PubMed]
  17. Feigin, V.L.; multiple collaborators. Global, regional, and national burden of neurological disorders during 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. The Lancet Neurology 2017, 16, 877–897. [Google Scholar] [CrossRef] [PubMed]
  18. Falk, E.B.; Hyde, L.W.; Mitchell, C.; Faul, J.; Gonzalez, R.; Heitzeg, M.M.; Keating, D.P.; Langa, K.M.; Martz, M.E.; Maslowsky, J.; Morrison, F.J.; Noll, D.C.; Patrick, M.E.; Pfeffer, F.T.; Reuter-Lorenz, P.A.; Thomason, M.E.; Davis-Kean, P.; Monk, C.S.; Schulenberg, J. What is a representative brain? Neuroscience meets population science. Proceedings of the National Academy of Sciences 2013, 110, 17615–17622. [Google Scholar] [CrossRef] [PubMed]
  19. Dotson, V.M.; Duarte, A. The importance of diversity in cognitive neuroscience. Annals of the New York Academy of Sciences 2019, 1464, 181–191. [Google Scholar] [CrossRef] [PubMed]
  20. Green, K.H.; Groep, I.H.V.D.; Brinke, L.W.T.; van der Cruijsen, R.; van Rossenberg, F.; Marroun, H.E. A perspective on enhancing representative samples in developmental human neuroscience: Connecting science to society. Frontiers in Integrative Neuroscience 2022, 16. [Google Scholar] [CrossRef]
  21. Siuly, S.; Zhang, Y. Medical Big Data: Neurological Diseases Diagnosis Through Medical Data Analysis. Data Science and Engineering 2016, 1, 54–64. [Google Scholar] [CrossRef]
  22. Alonso, S.G.; de la Torre-Díez, I.; Hamrioui, S.; López-Coronado, M.; Barreno, D.C.; Nozaleda, L.M.; Franco, M. Data Mining Algorithms and Techniques in Mental Health: A Systematic Review. Journal of Medical Systems 2018, 42. [Google Scholar] [CrossRef] [PubMed]
  23. Dipietro, L.; Gonzalez-Mego, P.; Ramos-Estebanez, C.; Zukowski, L.H.; Mikkilineni, R.; Rushmore, R.J.; Wagner, T. The evolution of Big Data in neuroscience and neurology. Journal of Big Data 2023, 10. [Google Scholar] [CrossRef] [PubMed]
  24. Li, R. Data Mining and Machine Learning Methods for Dementia Research. In Biomarkers for Alzheimer’s Disease Drug Development; Springer New York, 2018; pp. 363–370. [CrossRef]
  25. Baniya, B.; Athawale, S.V.; Choudhary, M.L.; Ram, N. Neurodegenerative Alzheimer’s Disease Disorders and Deep Learning Approaches. In Data Analysis for Neurodegenerative Disorders; Springer Nature Singapore, 2023; pp. 49–66. [CrossRef]
  26. Eschenburg, K.M.; Grabowski, T.J.; Haynor, D.R. Learning Cortical Parcellations Using Graph Neural Networks. Frontiers in Neuroscience 2021, 15. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, F.; Zhang, Y.; Rekik, I.; Massoud, Y.; Solé-Casals, J. Editorial: Graph learning for brain imaging. Frontiers in Neuroscience 2022, 16. [Google Scholar] [CrossRef] [PubMed]
  28. Qiu, W.; Ma, L.; Jiang, T.; Zhang, Y. Unrevealing Reliable Cortical Parcellation of Individual Brains Using Resting-State Functional Magnetic Resonance Imaging and Masked Graph Convolutions. Frontiers in Neuroscience 2022, 16. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, X.; Yang, Y.; Kuai, H.; Chen, J.; Huang, J.; Liang, P.; Zhong, N. Systematic Fusion of Multi-Source Cognitive Networks With Graph Learning - A Study on Fronto-Parietal Network. Frontiers in Neuroscience 2022, 16. [Google Scholar] [CrossRef] [PubMed]
  30. Kurucu, M.C.; Rekik, I. Graph neural network based unsupervised influential sample selection for brain multigraph population fusion. Computerized Medical Imaging and Graphics 2023, 108, 102274. [Google Scholar] [CrossRef] [PubMed]
  31. Bessadok, A.; Mahjoub, M.A.; Rekik, I. Graph Neural Networks in Network Neuroscience. IEEE Transactions on Pattern Analysis and Machine Intelligence 2023, 45, 5833–5848. [Google Scholar] [CrossRef] [PubMed]
  32. Najarro, E.; Sudhakaran, S.; Risi, S. Towards Self-Assembling Artificial Neural Networks through Neural Developmental Programs, 2023. [CrossRef]
  33. Brodmann, K. Vergleichende Lokalisationslehre der Großhirnrinde : in ihren Prinzipien dargestellt auf Grund des Zellenbaues; Barth, 1909.
  34. Ferris, C.F.; Cai, X.; Qiao, J.; Switzer, B.; Baun, J.; Morrison, T.; Iriah, S.; Madularu, D.; Sinkevicius, K.W.; Kulkarni, P. Life without a brain: Neuroradiological and behavioral evidence of neuroplasticity necessary to sustain brain function in the face of severe hydrocephalus. Scientific Reports 2019, 9. [Google Scholar] [CrossRef]
  35. Miller, A. The Lobotomy Patient. A Decade Later. Canadian Medical Association, 2016. [Google Scholar]
  36. Dickman, H.; Fletke, K.; Redfern, R.E. Prolonged unassisted survival in an infant with anencephaly. BMJ Case Reports, 2016. [Google Scholar] [CrossRef]
  37. Nagappan, P.G.; Chen, H.; Wang, D.Y. Neuroregeneration and plasticity: a review of the physiological mechanisms for achieving functional recovery postinjury. Military Medical Research 2020, 7. [Google Scholar] [CrossRef]
  38. Lim, S.B.; Louie, D.R.; Peters, S.; Liu-Ambrose, T.; Boyd, L.A.; Eng, J.J. Brain activity during real-time walking and with walking interventions after stroke: a systematic review. Journal of NeuroEngineering and Rehabilitation 2021, 18. [Google Scholar] [CrossRef] [PubMed]
  39. Bigler, E.D. Structural Image Analysis of the Brain in Neuropsychology Using Magnetic Resonance Imaging (MRI) Techniques. Neuropsychology Review 2015, 25, 224–249. [Google Scholar] [CrossRef] [PubMed]
  40. Silva, M.A.; See, A.P.; Essayed, W.I.; Golby, A.J.; Tie, Y. Challenges and techniques for presurgical brain mapping with functional MRI. NeuroImage: Clinical 2018, 17, 794–803. [Google Scholar] [CrossRef] [PubMed]
  41. Moghimi, P.; Dang, A.T.; Do, Q.; Netoff, T.I.; Lim, K.O.; Atluri, G. Evaluation of functional MRI-based human brain parcellation: a review. Journal of Neurophysiology 2022, 128, 197–217. [Google Scholar] [CrossRef] [PubMed]
  42. Hagmann, P.; Kurant, M.; Gigandet, X.; Thiran, P.; Wedeen, V.J.; Meuli, R.; Thiran, J.P. Mapping Human Whole-Brain Structural Networks with Diffusion MRI. PLoS ONE 2007, 2, e597. [Google Scholar] [CrossRef] [PubMed]
  43. Sagar, S.; Rick, J.; Chandra, A.; Yagnik, G.; Aghi, M.K. Functional brain mapping: overview of techniques and their application to neurosurgery. Neurosurgical Review 2018, 42, 639–647. [Google Scholar] [CrossRef] [PubMed]
  44. Alkemade, A.; Bazin, P.L.; Balesar, R.; Pine, K.; Kirilina, E.; Möller, H.E.; Trampel, R.; Kros, J.M.; Keuken, M.C.; Bleys, R.L.A.W.; Swaab, D.F.; Herrler, A.; Weiskopf, N.; Forstmann, B.U. A unified 3D map of microscopic architecture and MRI of the human brain. Science Advances 2022, 8. [Google Scholar] [CrossRef]
  45. Amunts, K.; Mohlberg, H.; Bludau, S.; Zilles, K. Julich-Brain: A 3D probabilistic atlas of the human brain’s cytoarchitecture. Science 2020, 369, 988–992. [Google Scholar] [CrossRef]
  46. Borys, D.; Kijonka, M.; Psiuk-Maksymowicz, K.; Gorczewski, K.; Zarudzki, L.; Sokol, M.; Swierniak, A. Non-parametric MRI Brain Atlas for the Polish Population. Frontiers in Neuroinformatics 2021, 15. [Google Scholar] [CrossRef]
  47. Zhao, B.; Li, T.; Li, Y.; Fan, Z.; Xiong, D.; Wang, X.; Gao, M.; Smith, S.M.; Zhu, H. An atlas of trait associations with resting-state and task-evoked human brain functional organizations in the UK Biobank. Imaging Neuroscience 2023, 1, 1–23. [Google Scholar] [CrossRef]
  48. Tan, L.H.; Chan, A.H.D.; Kay, P.; Khong, P.L.; Yip, L.K.C.; Luke, K.K. Language affects patterns of brain activation associated with perceptual decision. Proceedings of the National Academy of Sciences 2008, 105, 4004–4009. [Google Scholar] [CrossRef] [PubMed]
  49. Toro, R.; Fox, P.T.; Paus, T. Functional Coactivation Map of the Human Brain. Cerebral Cortex 2008, 18, 2553–2559. [Google Scholar] [CrossRef] [PubMed]
  50. Laird, A.R.; Eickhoff, S.B.; Rottschy, C.; Bzdok, D.; Ray, K.L.; Fox, P.T. Networks of task co-activations. NeuroImage 2013, 80, 505–514. [Google Scholar] [CrossRef] [PubMed]
  51. Shibata, K.; Watanabe, T.; Kawato, M.; Sasaki, Y. Differential Activation Patterns in the Same Brain Region Led to Opposite Emotional States. PLOS Biology 2016, 14, e1002546. [Google Scholar] [CrossRef] [PubMed]
  52. Nakuci, J.; Yeon, J.; Kim, J.H.; Kim, S.P.; Rahnev, D. Multiple brain activation patterns for the same task 2023. [CrossRef]
  53. Baek, C.Y.; Kim, H.D.; Yoo, D.Y.; Kang, K.Y.; Lee, J.W. Change in activity patterns in the prefrontal cortex in different phases during the dual-task walking in older adults. Journal of NeuroEngineering and Rehabilitation 2023, 20. [Google Scholar] [CrossRef] [PubMed]
  54. Buckner, R.L.; DiNicola, L.M. The brain’s default network: updated anatomy, physiology and evolving insights. Nature Reviews Neuroscience 2019, 20, 593–608. [Google Scholar] [CrossRef] [PubMed]
  55. Smallwood, J.; Bernhardt, B.C.; Leech, R.; Bzdok, D.; Jefferies, E.; Margulies, D.S. The default mode network in cognition: a topographical perspective. Nature Reviews Neuroscience 2021, 22, 503–513. [Google Scholar] [CrossRef] [PubMed]
  56. Buckner, R.L. The brain’s default network: origins and implications for the study of psychosis. Dialogues in Clinical Neuroscience 2013, 15, 351–358. [Google Scholar] [CrossRef] [PubMed]
  57. Siddiqi, S.H.; Kording, K.P.; Parvizi, J.; Fox, M.D. Causal mapping of human brain function. Nature Reviews Neuroscience 2022, 23, 361–375. [Google Scholar] [CrossRef]
  58. Finger, S.; Almli, C. Brain damage and neuroplasticity: Mechanisms of recovery or development? Brain Research Reviews 1985, 10, 177–186. [Google Scholar] [CrossRef]
  59. Grafman, J. Conceptualizing functional neuroplasticity. Journal of Communication Disorders 2000, 33, 345–356. [Google Scholar] [CrossRef]
  60. Fuchs, E.; Flügge, G. Adult Neuroplasticity: More Than 40 Years of Research. Neural Plasticity 2014, 2014, 1–10. [Google Scholar] [CrossRef]
  61. Kolb, B.; Gibb, R. Brain Plasticity and Behaviour in the Developing Brain. Journal of the Canadian Academy of Child and Adolescent Psychiatry 2011. [Google Scholar]
  62. Park, D.C.; Bischof, G.N. The aging mind: neuroplasticity in response to cognitive training. Dialogues in Clinical Neuroscience 2013, 15, 109–119. [Google Scholar] [CrossRef] [PubMed]
  63. Viel, T.A.; Toricelli, M.; Pereira, A.R.; Abrao, G.S.; Malerba, H.N.; Maia, J.; Buck, H.S. Mechanisms of neuroplasticity and brain degeneration: strategies for protection during the aging process. Neural Regeneration Research 2021, 16, 58. [Google Scholar] [CrossRef] [PubMed]
  64. Bennett, S.H.; Kirby, A.J.; Finnerty, G.T. Rewiring the connectome: Evidence and effects. Neuroscience and Biobehavioral Reviews 2018, 88, 51–62. [Google Scholar] [CrossRef]
  65. Seven, Y.B.; Mitchell, G.S. Mechanisms of compensatory plasticity for respiratory motor neuron death. Respiratory Physiology and Neurobiology 2019, 265, 32–39. [Google Scholar] [CrossRef]
  66. Hill, N.L.; Kolanowski, A.M.; Gill, D.J. Plasticity in Early Alzheimer Disease. Topics in Geriatric Rehabilitation 2011, 27, 257–267. [Google Scholar] [CrossRef]
  67. Meyer, J.S.; Obara, K.; Muramatsu, K. Diaschisis. Neurological Research 1993, 15, 362–366. [Google Scholar] [CrossRef]
  68. Elmore, S. Apoptosis: A Review of Programmed Cell Death. Toxicologic Pathology 2007, 35, 495–516. [Google Scholar] [CrossRef]
  69. Hutchins, J.B.; Barger, S.W. Why neurons die: Cell death in the nervous system. The Anatomical Record 1998, 253, 79–90. [Google Scholar] [CrossRef]
  70. Yamaguchi, Y.; Miura, M. Programmed Cell Death in Neurodevelopment. Developmental Cell 2015, 32, 478–490. [Google Scholar] [CrossRef] [PubMed]
  71. Dekkers, M.P.; Nikoletopoulou, V.; Barde, Y.A. Death of developing neurons: New insights and implications for connectivity. Journal of Cell Biology 2013, 203, 385–393. [Google Scholar] [CrossRef] [PubMed]
  72. Kristiansen, M.; Ham, J. Programmed cell death during neuronal development: the sympathetic neuron model. Cell Death and Differentiation 2014, 21, 1025–1035. [Google Scholar] [CrossRef] [PubMed]
  73. Finger, S. Chapter 51 Recovery of function. In Handbook of Clinical Neurology; Elsevier, 2009; pp. 833–841. [CrossRef]
  74. Fricker, M.; Tolkovsky, A.M.; Borutaite, V.; Coleman, M.; Brown, G.C. Neuronal Cell Death. Physiological Reviews 2018, 98, 813–880. [Google Scholar] [CrossRef] [PubMed]
  75. Pettmann, B.; Henderson, C.E. Neuronal Cell Death. Neuron 1998, 20, 633–647. [Google Scholar] [CrossRef]
  76. Lowe, S.W.; Lin, A.W. Apoptosis in cancer. Carcinogenesis 2000, 21, 485–495. [Google Scholar] [CrossRef] [PubMed]
  77. Wong, R.S. Apoptosis in cancer: from pathogenesis to treatment. Journal of Experimental and Clinical Cancer Research 2011, 30. [Google Scholar] [CrossRef]
  78. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nature Reviews Clinical Oncology 2020, 17, 395–417. [Google Scholar] [CrossRef]
  79. Chi, H.; Chang, H.Y.; Sang, T.K. Neuronal Cell Death Mechanisms in Major Neurodegenerative Diseases. International Journal of Molecular Sciences 2018, 19, 3082. [Google Scholar] [CrossRef]
  80. Goel, P.; Chakrabarti, S.; Goel, K.; Bhutani, K.; Chopra, T.; Bali, S. Neuronal cell death mechanisms in Alzheimer’s disease: An insight. Frontiers in Molecular Neuroscience 2022, 15. [Google Scholar] [CrossRef]
  81. Zafarlotfi, S.; Quadri, M.; Borodovsky, J. Understanding Brain Damage and Sleep Apnea: A Review. Health Outcomes Research in Medicine 2010, 1, e103–e110. [Google Scholar] [CrossRef]
  82. Lin, S.Y.; Chen, W.; Harnod, T.; Lin, C.L.; Hsu, W.H.; Lin, C.C.; Chang, Y.L.; Wang, I.K.; Kao, C.H. Sleep apnea and risk of traumatic brain injury and associated mortality and healthcare costs: a population-based cohort study. Annals of Translational Medicine 2019, 7, 644–644. [Google Scholar] [CrossRef] [PubMed]
  83. Giustini, A.; Pistarini, C.; Pisoni, C. Traumatic and nontraumatic brain injury. In Neurological Rehabilitation; Elsevier, 2013; pp. 401–409. [CrossRef]
  84. Hohmann, U.; Dehghani, F.; Hohmann, T. Assessment of Neuronal Damage in Brain Slice Cultures Using Machine Learning Based on Spatial Features. Frontiers in Neuroscience 2021, 15. [Google Scholar] [CrossRef]
  85. Mamere, A.; Saraiva, L.; Matos, A.; Carneiro, A.; Santos, A. Evaluation of Delayed Neuronal and Axonal Damage Secondary to Moderate and Severe Traumatic Brain Injury Using Quantitative MR Imaging Techniques. American Journal of Neuroradiology 2009, 30, 947–952. [Google Scholar] [CrossRef]
  86. Majdan, M.; Plancikova, D.; Brazinova, A.; Rusnak, M.; Nieboer, D.; Feigin, V.; Maas, A. Epidemiology of traumatic brain injuries in Europe: a cross-sectional analysis. The Lancet Public Health 2016, 1, e76–e83. [Google Scholar] [CrossRef]
  87. Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano, A.M.; Rosenfeld, J.V.; Park, K.B. Estimating the global incidence of traumatic brain injury. Journal of Neurosurgery 2019, 130, 1080–1097. [Google Scholar] [CrossRef] [PubMed]
  88. Goldman, L.; Siddiqui, E.M.; Khan, A.; Jahan, S.; Rehman, M.U.; Mehan, S.; Sharma, R.; Budkin, S.; Kumar, S.N.; Sahu, A.; Kumar, M.; Vaibhav, K. Understanding Acquired Brain Injury: A Review. Biomedicines 2022, 10, 2167. [Google Scholar] [CrossRef]
  89. Rogers, J.M.; Read, C.A. Psychiatric comorbidity following traumatic brain injury. Brain Injury 2007, 21, 1321–1333. [Google Scholar] [CrossRef] [PubMed]
  90. Hammond, F.M.; Corrigan, J.D.; Ketchum, J.M.; Malec, J.F.; Dams-O’Connor, K.; Hart, T.; Novack, T.A.; Bogner, J.; Dahdah, M.N.; Whiteneck, G.G. Prevalence of Medical and Psychiatric Comorbidities Following Traumatic Brain Injury. Journal of Head Trauma Rehabilitation 2019, 34, E1–E10. [Google Scholar] [CrossRef]
  91. Naumenko, Y.; Yuryshinetz, I.; Zabenko, Y.; Pivneva, T. Mild traumatic brain injury as a pathological process. Heliyon 2023, 9, e18342. [Google Scholar] [CrossRef] [PubMed]
  92. Dean, P.J.A.; Sterr, A. Long-term effects of mild traumatic brain injury on cognitive performance. Frontiers in Human Neuroscience 2013, 7. [Google Scholar] [CrossRef] [PubMed]
  93. Bramlett, H.M.; Dietrich, W.D. Long-Term Consequences of Traumatic Brain Injury: Current Status of Potential Mechanisms of Injury and Neurological Outcomes. Journal of Neurotrauma 2015, 32, 1834–1848. [Google Scholar] [CrossRef] [PubMed]
  94. Danna-Dos-Santos, A.; Mohapatra, S.; Santos, M.; Degani, A.M. Long-term effects of mild traumatic brain injuries to oculomotor tracking performances and reaction times to simple environmental stimuli. Scientific Reports 2018, 8. [Google Scholar] [CrossRef] [PubMed]
  95. McKee, A.C.; Cantu, R.C.; Nowinski, C.J.; Hedley-Whyte, E.T.; Gavett, B.E.; Budson, A.E.; Santini, V.E.; Lee, H.S.; Kubilus, C.A.; Stern, R.A. Chronic Traumatic Encephalopathy in Athletes: Progressive Tauopathy After Repetitive Head Injury. Journal of Neuropathology and Experimental Neurology 2009, 68, 709–735. [Google Scholar] [CrossRef] [PubMed]
  96. Sudhakar, S.K.; Sridhar, S.; Char, S.; Pandya, K.; Mehta, K. Prevalence of comorbidities post mild traumatic brain injuries: a traumatic brain injury model systems study. Frontiers in Human Neuroscience 2023, 17. [Google Scholar] [CrossRef] [PubMed]
  97. Das, A.S.; Vicenty-Padilla, J.C.; Chua, M.M.; Jeelani, Y.; Snider, S.B.; Regenhardt, R.W.; Al-Mufti, F.; Du, R.; Izzy, S. Cerebrovascular injuries in traumatic brain injury. Clinical Neurology and Neurosurgery 2022, 223, 107479. [Google Scholar] [CrossRef] [PubMed]
  98. Rashid, B. Mechanical Characterization of Brain Tissue in Compression, Tension and Shear Under Dynamic Conditions. PhD thesis, School of Mechanical and Materials Engineering, University College Dublin, 2012.
  99. Accessed: 09-10-2023.
  100. Mokri, B. The Monro-Kellie hypothesis: Applications in CSF volume depletion. Neurology 2001, 56, 1746–1748. [Google Scholar] [CrossRef] [PubMed]
  101. Kalisvaart, A.C.J.; Wilkinson, C.M.; Gu, S.; Kung, T.F.C.; Yager, J.; Winship, I.R.; van Landeghem, F.K.H.; Colbourne, F. An update to the Monro–Kellie doctrine to reflect tissue compliance after severe ischemic and hemorrhagic stroke. Scientific Reports 2020, 10. [Google Scholar] [CrossRef]
  102. Cruz-Haces, M.; Tang, J.; Acosta, G.; Fernandez, J.; Shi, R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Translational Neurodegeneration 2017, 6. [Google Scholar] [CrossRef]
  103. Graham, N.S.; Sharp, D.J. Understanding neurodegeneration after traumatic brain injury: from mechanisms to clinical trials in dementia. Journal of Neurology, Neurosurgery and Psychiatry 2019, 90, 1221–1233. [Google Scholar] [CrossRef]
  104. Graham, N.S.N.; Jolly, A.; Zimmerman, K.; Bourke, N.J.; Scott, G.; Cole, J.H.; Schott, J.M.; Sharp, D.J. Diffuse axonal injury predicts neurodegeneration after moderate–severe traumatic brain injury. Brain 2020, 143, 3685–3698. [Google Scholar] [CrossRef]
  105. Dodd, W.S.; Panther, E.J.; Pierre, K.; Hernandez, J.S.; Patel, D.; Lucke-Wold, B. Traumatic Brain Injury and Secondary Neurodegenerative Disease. Trauma Care 2022, 2, 510–522. [Google Scholar] [CrossRef]
  106. Li, X.Y.; Feng, D.F. Diffuse axonal injury: Novel insights into detection and treatment. Journal of Clinical Neuroscience 2009, 16, 614–619. [Google Scholar] [CrossRef]
  107. Vo, D.T.; Phan, C.C.; Le, H.G.N.; Vo, T.P.; Mai, U.T.T.; Le, H.K.; Ha, T.B.T. Diffuse axonal injury: a case report and MRI findings. Radiology Case Reports 2022, 17, 91–94. [Google Scholar] [CrossRef]
  108. Markl, M.; Leupold, J. Gradient echo imaging. Journal of Magnetic Resonance Imaging 2012, 35, 1274–1289. [Google Scholar] [CrossRef]
  109. Haller, S.; Haacke, E.M.; Thurnher, M.M.; Barkhof, F. Susceptibility-weighted Imaging: Technical Essentials and Clinical Neurologic Applications. Radiology 2021, 299, 3–26. [Google Scholar] [CrossRef]
  110. Skarupa, D.J.; Khan, M.; Hsu, A.; Madbak, F.G.; Ebler, D.J.; Yorkgitis, B.; Rahmathulla, G.; Alcindor, D.; Joseph, B. Trends in civilian penetrating brain injury: A review of 26, 871 patients. The American Journal of Surgery 2019, 218, 255–260. [Google Scholar] [CrossRef]
  111. Günther, M.; Dahlberg, M.; Rostami, A.; Azadali, A.; Arborelius, U.P.; Linder, F.; Rostami, E. Incidence, Demographics, and Outcomes of Penetrating Trauma in Sweden During the Past Decade. Frontiers in Neurology 2021, 12. [Google Scholar] [CrossRef]
  112. Vakil, M.T.; Singh, A.K. A review of penetrating brain trauma: epidemiology, pathophysiology, imaging assessment, complications, and treatment. Emergency Radiology 2017, 24, 301–309. [Google Scholar] [CrossRef]
  113. Raymont, V.; Salazar, A.M.; Lipsky, R.; Goldman, D.; Tasick, G.; Grafman, J. Correlates of posttraumatic epilepsy 35 years following combat brain injury. Neurology 2010, 75, 224–229. [Google Scholar] [CrossRef]
  114. Accessed: 11-10-2023.
  115. Enam, S.; Kazim, S.; Shamim, M.; Tahir, M.; Waheed, S. Management of penetrating brain injury. Journal of Emergencies, Trauma, and Shock 2011, 4, 395. [Google Scholar] [CrossRef] [PubMed]
  116. Wong, C.P. Current topic: Incidence, aetiology, and outcome of non-traumatic coma: a population based study. Archives of Disease in Childhood 2001, 84, 193–199. [Google Scholar] [CrossRef]
  117. Sarrazin, J.L.; Bonneville, F.; Martin-Blondel, G. Brain infections. Diagnostic and Interventional Imaging 2012, 93, 473–490. [Google Scholar] [CrossRef]
  118. Abdullahi, A.M.; Sarmast, S.T.; Singh, R. Molecular Biology and Epidemiology of Neurotropic Viruses. Cureus 2020. [Google Scholar] [CrossRef]
  119. Godkhindi, V.M.; Monappa, V.; Kairanna, N.V.; Sharma, S.; Vasudevan, G.; Hebbar, K.D. Brain infections that mimic malignancy. Diagnostic Histopathology 2022, 28, 456–466. [Google Scholar] [CrossRef]
  120. Sekino, N.; Selim, M.; Shehadah, A. Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments. Journal of Neuroinflammation 2022, 19. [Google Scholar] [CrossRef]
  121. Lv, S.; Zhang, Y.; Steinmann, P.; Zhou, X.N.; Utzinger, J. Helminth Infections of the Central Nervous System Occurring in Southeast Asia and the Far East. In Important Helminth Infections in Southeast Asia: Diversity and Potential for Control and Elimination, Part A; Elsevier, 2010; pp. 351–408. [CrossRef]
  122. Owusu-Agyei, A.K.; Nabarro, L. Helminth infections and differentials of eosinophilia. Medicine 2021, 49, 766–769. [Google Scholar] [CrossRef]
  123. Accessed: 19-10-2023.
  124. Shadmani, G.; Simkins, T.J.; Assadsangabi, R.; Apperson, M.; Hacein-Bey, L.; Raslan, O.; Ivanovic, V. Autoimmune diseases of the brain, imaging and clinical review. The Neuroradiology Journal 2021, 35, 152–169. [Google Scholar] [CrossRef]
  125. McGlasson, S.; Wiseman, S.; Wardlaw, J.; Dhaun, N.; Hunt, D.P.J. Neurological Disease in Lupus: Toward a Personalized Medicine Approach. Frontiers in Immunology 2018, 9. [Google Scholar] [CrossRef]
  126. Kayser, M.S.; Dalmau, J. The Emerging Link Between Autoimmune Disorders and Neuropsychiatric Disease. Journal of Neuropsychiatry 2011, 23, 90–97. [Google Scholar] [CrossRef]
  127. Wiethoff, S.; Houlden, H. Neurodegeneration with brain iron accumulation. In Handbook of Clinical Neurology; Elsevier, 2018; pp. 157–166. [CrossRef]
  128. Buyukcoban, S.; Arici, M.A.; Koca, U.; Kalkan, S. A Case Report of Toxic Brain Syndrome Caused by Methyl Bromide. Turkish Journal of Anesthesia and Reanimation 2015, 43, 134–137. [Google Scholar] [CrossRef]
  129. Luvisetto, S. Botulinum Neurotoxins in Central Nervous System: An Overview from Animal Models to Human Therapy. Toxins 2021, 13, 751. [Google Scholar] [CrossRef]
  130. Weis, S.; Büttner, A. Neurotoxicology and drug-related disorders. In Handbook of Clinical Neurology; Elsevier, 2018; pp. 181–192. [CrossRef]
  131. Kim, Y.; Kim, J.W. Toxic Encephalopathy. Safety and Health at Work 2012, 3, 243–256. [Google Scholar] [CrossRef]
  132. Moratalla, R.; Khairnar, A.; Simola, N.; Granado, N.; García-Montes, J.R.; Porceddu, P.F.; Tizabi, Y.; Costa, G.; Morelli, M. Amphetamine-related drugs neurotoxicity in humans and in experimental animals: Main mechanisms. Progress in Neurobiology 2017, 155, 149–170. [Google Scholar] [CrossRef]
  133. Cunha-Oliveira, T.; Rego, A.C.; Oliveira, C.R. Cellular and molecular mechanisms involved in the neurotoxicity of opioid and psychostimulant drugs. Brain Research Reviews 2008, 58, 192–208. [Google Scholar] [CrossRef]
  134. Guennec, L.L.; Marois, C.; Demeret, S.; Wijdicks, E.; Weiss, N. Toxic-metabolic encephalopathy in adults: Critical discussion and pragmatical diagnostic approach. Revue Neurologique 2022, 178, 93–104. [Google Scholar] [CrossRef]
  135. Weis, S.; Büttner, A. Nutritional and systemic metabolic disorders. In Handbook of Clinical Neurology; Elsevier, 2018; pp. 167–173. [CrossRef]
  136. Greene-Schloesser, D.; Robbins, M.E.; Peiffer, A.M.; Shaw, E.G.; Wheeler, K.T.; Chan, M.D. Radiation-induced brain injury: A review. Frontiers in Oncology 2012, 2. [Google Scholar] [CrossRef]
  137. Belka, C.; Budach, W.; Kortmann, R.D.; Bamberg, M. Radiation induced CNS toxicity – molecular and cellular mechanisms. British Journal of Cancer 2001, 85, 1233–1239. [Google Scholar] [CrossRef] [PubMed]
  138. Furuse, M.; Nonoguchi, N.; Kawabata, S.; Miyatake, S.I.; Kuroiwa, T. Delayed brain radiation necrosis: pathological review and new molecular targets for treatment. Medical Molecular Morphology 2015, 48, 183–190. [Google Scholar] [CrossRef] [PubMed]
  139. Kim, J.H.; Brown, S.L.; Jenrow, K.A.; Ryu, S. Mechanisms of radiation-induced brain toxicity and implications for future clinical trials. Journal of Neuro-Oncology 2008, 87, 279–286. [Google Scholar] [CrossRef]
  140. Ali, F.S.; Arevalo, O.; Zorofchian, S.; Patrizz, A.; Riascos, R.; Tandon, N.; Blanco, A.; Ballester, L.Y.; Esquenazi, Y. Cerebral Radiation Necrosis: Incidence, Pathogenesis, Diagnostic Challenges, and Future Opportunities. Current Oncology Reports 2019, 21. [Google Scholar] [CrossRef] [PubMed]
  141. Lawrence, Y.R.; Li, X.A.; el Naqa, I.; Hahn, C.A.; Marks, L.B.; Merchant, T.E.; Dicker, A.P. Radiation Dose-Volume Effects in the Brain. International Journal of Radiation Oncology, Biology and Physics 2010, 76, S20–S27. [Google Scholar] [CrossRef] [PubMed]
  142. Smith, E.J.; Naik, A.; Shaffer, A.; Goel, M.; Krist, D.T.; Liang, E.; Furey, C.G.; Miller, W.K.; Lawton, M.T.; Barnett, D.H.; Weis, B.; Rizk, A.; Smith, R.S.; Hassaneen, W. Differentiating radiation necrosis from tumor recurrence: a systematic review and diagnostic meta-analysis comparing imaging modalities. Journal of Neuro-Oncology 2023, 162, 15–23. [Google Scholar] [CrossRef] [PubMed]
  143. Kenney, K.; Amyot, F.; Haber, M.; Pronger, A.; Bogoslovsky, T.; Moore, C.; Diaz-Arrastia, R. Cerebral Vascular Injury in Traumatic Brain Injury. Experimental Neurology 2016, 275, 353–366. [Google Scholar] [CrossRef] [PubMed]
  144. Murphy, S.J.; Werring, D.J. Stroke: causes and clinical features. Medicine 2020, 48, 561–566. [Google Scholar] [CrossRef] [PubMed]
  145. Grysiewicz, R.A.; Thomas, K.; Pandey, D.K. Epidemiology of Ischemic and Hemorrhagic Stroke: Incidence, Prevalence, Mortality, and Risk Factors. Neurologic Clinics 2008, 26, 871–895. [Google Scholar] [CrossRef]
  146. Silasi, G.; Murphy, T.H. Stroke and the Connectome: How Connectivity Guides Therapeutic Intervention. Neuron 2014, 83, 1354–1368. [Google Scholar] [CrossRef]
  147. Baldassarre, A.; Ramsey, L.E.; Siegel, J.S.; Shulman, G.L.; Corbetta, M. Brain connectivity and neurological disorders after stroke. Current Opinion in Neurology 2016, 29, 706–713. [Google Scholar] [CrossRef]
  148. Jiang, L.; Xu, H.; Yu, C. Brain Connectivity Plasticity in the Motor Network after Ischemic Stroke. Neural Plasticity 2013, 2013, 1–11. [Google Scholar] [CrossRef]
  149. Li, W.; Li, Y.; Zhu, W.; Chen, X. Changes in brain functional network connectivity after stroke. Neural Regeneration Research 2014, 9, 51. [Google Scholar] [CrossRef] [PubMed]
  150. Desowska, A.; Turner, D.L. Dynamics of brain connectivity after stroke. Reviews in the Neurosciences 2019, 30, 605–623. [Google Scholar] [CrossRef] [PubMed]
  151. Hall, G.R.; Kaiser, M.; Farr, T.D. Functional Connectivity Change in Response to Stroke Is Comparable Across Species: From Mouse to Man. Stroke 2021, 52, 2961–2963. [Google Scholar] [CrossRef] [PubMed]
  152. McFaline-Figueroa, J.R.; Lee, E.Q. Brain Tumors. The American Journal of Medicine 2018, 131, 874–882. [Google Scholar] [CrossRef] [PubMed]
  153. DeAngelis, L.M. Brain Tumors. New England Journal of Medicine 2001, 344, 114–123. [Google Scholar] [CrossRef] [PubMed]
  154. Herholz, K.; Langen, K.J.; Schiepers, C.; Mountz, J.M. Brain Tumors. Seminars in Nuclear Medicine 2012, 42, 356–370. [Google Scholar] [CrossRef] [PubMed]
  155. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathologica 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed]
  156. Lapointe, S.; Perry, A.; Butowski, N.A. Primary brain tumours in adults. The Lancet 2018, 392, 432–446. [Google Scholar] [CrossRef] [PubMed]
  157. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.; Simjee, S. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pacific Journal of Cancer Prevention 2017, 18. [Google Scholar] [CrossRef]
  158. de Oliveira Costa, L.L.; Brandão, E.C.; Segundo, L.M.D.B.M. Atualização em epilepsia. Revista de Medicina 2020, 99, 170–181. [Google Scholar] [CrossRef]
  159. Scharfman, H.E. The neurobiology of epilepsy. Current Neurology and Neuroscience Reports 2007, 7, 348–354. [Google Scholar] [CrossRef] [PubMed]
  160. Beghi, E. The Epidemiology of Epilepsy. Neuroepidemiology 2019, 54, 185–191. [Google Scholar] [CrossRef]
  161. Stafstrom, C.E.; Carmant, L. Seizures and Epilepsy: An Overview for Neuroscientists. Cold Spring Harbor Perspectives in Medicine 2015, 5, a022426–a022426. [Google Scholar] [CrossRef]
  162. Banerjee, P.N.; Filippi, D.; Hauser, W.A. The descriptive epidemiology of epilepsy—A review. Epilepsy Research 2009, 85, 31–45. [Google Scholar] [CrossRef] [PubMed]
  163. Staley, K. Epileptic Neurons Go Wireless. Science 2004, 305, 482–483. [Google Scholar] [CrossRef]
  164. Duncan, J.S.; Sander, J.W.; Sisodiya, S.M.; Walker, M.C. Adult epilepsy. The Lancet 2006, 367, 1087–1100. [Google Scholar] [CrossRef] [PubMed]
  165. Stefan, H.; da Silva, F.H.L. Epileptic Neuronal Networks: Methods of Identification and Clinical Relevance. Frontiers in Neurology 2013, 4. [Google Scholar] [CrossRef] [PubMed]
  166. Ren, X.; Brodovskaya, A.; Hudson, J.L.; Kapur, J. Connectivity and Neuronal Synchrony during Seizures. The Journal of Neuroscience 2021, 41, 7623–7635. [Google Scholar] [CrossRef]
  167. Schependom, J.V.; D’haeseleer, M. Advances in Neurodegenerative Diseases. Journal of Clinical Medicine 2023, 12, 1709. [Google Scholar] [CrossRef]
  168. Davenport, F.; Gallacher, J.; Kourtzi, Z.; Koychev, I.; Matthews, P.M.; Oxtoby, N.P.; Parkes, L.M.; Priesemann, V.; Rowe, J.B.; Smye, S.W.; Zetterberg, H. Neurodegenerative disease of the brain: a survey of interdisciplinary approaches. Journal of The Royal Society Interface 2023, 20. [Google Scholar] [CrossRef]
  169. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harbor Perspectives in Biology 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed]
  170. Amor, S.; Puentes, F.; Baker, D.; Valk, P.V.D. Inflammation in neurodegenerative diseases. Immunology 2010, 129, 154–169. [Google Scholar] [CrossRef] [PubMed]
  171. Arvanitakis, Z.; Shah, R.C.; Bennett, D.A. Diagnosis and Management of Dementia: Review. JAMA 2019, 322, 1589. [Google Scholar] [CrossRef] [PubMed]
  172. Livingston, G.; Huntley, J.; Sommerlad, A.; Ames, D.; Ballard, C.; Banerjee, S.; Brayne, C.; Burns, A.; Cohen-Mansfield, J.; Cooper, C.; Costafreda, S.G.; Dias, A.; Fox, N.; Gitlin, L.N.; Howard, R.; Kales, H.C.; Kivimäki, M.; Larson, E.B.; Ogunniyi, A.; Orgeta, V.; Ritchie, K.; Rockwood, K.; Sampson, E.L.; Samus, Q.; Schneider, L.S.; Selbæk, G.; Teri, L.; Mukadam, N. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. The Lancet 2020, 396, 413–446. [Google Scholar] [CrossRef]
  173. Checkoway, H.; Lundin, J.I.; Kelada, S.N.
  174. Niu, H.; Álvarez-Álvarez, I.; Guillén-Grima, F.; Aguinaga-Ontoso, I. Prevalencia e incidencia de la enfermedad de Alzheimer en Europa: metaanálisis. Neurología 2017, 32, 523–532. [Google Scholar] [CrossRef] [PubMed]
  175. Bartzokis, G. Alzheimer’s disease as homeostatic responses to age-related myelin breakdown. Neurobiology of Aging 2011, 32, 1341–1371. [Google Scholar] [CrossRef] [PubMed]
  176. Depp, C.; Sun, T.; Sasmita, A.O.; Spieth, L.; Berghoff, S.A.; Nazarenko, T.; Overhoff, K.; Steixner-Kumar, A.A.; Subramanian, S.; Arinrad, S.; Ruhwedel, T.; Möbius, W.; Göbbels, S.; Saher, G.; Werner, H.B.; Damkou, A.; Zampar, S.; Wirths, O.; Thalmann, M.; Simons, M.; Saito, T.; Saido, T.; Krueger-Burg, D.; Kawaguchi, R.; Willem, M.; Haass, C.; Geschwind, D.; Ehrenreich, H.; Stassart, R.; Nave, K.A. Myelin dysfunction drives amyloid-β deposition in models of Alzheimer’s disease. Nature 2023, 618, 349–357. [Google Scholar] [CrossRef] [PubMed]
  177. Cain, A.; Taga, M.; McCabe, C.; Green, G.S.; Hekselman, I.; White, C.C.; Lee, D.I.; Gaur, P.; Rozenblatt-Rosen, O.; Zhang, F.; Yeger-Lotem, E.; Bennett, D.A.; Yang, H.S.; Regev, A.; Menon, V.; Habib, N.; Jager, P.L.D. Multicellular communities are perturbed in the aging human brain and Alzheimer’s disease. Nature Neuroscience 2023, 26, 1267–1280. [Google Scholar] [CrossRef] [PubMed]
  178. Stam, C.; Jones, B.; Nolte, G.; Breakspear, M.; Scheltens, P. Small-World Networks and Functional Connectivity in Alzheimer’s Disease. Cerebral Cortex 2006, 17, 92–99. [Google Scholar] [CrossRef]
  179. Yu, M.; Sporns, O.; Saykin, A.J. The human connectome in Alzheimer disease — relationship to biomarkers and genetics. Nature Reviews Neurology 2021, 17, 545–563. [Google Scholar] [CrossRef]
  180. Mårtensson, G.; Pereira, J.B.; Mecocci, P.; Vellas, B.; Tsolaki, M.; Kłoszewska, I.; Soininen, H.; Lovestone, S.; Simmons, A.; Volpe, G.; Westman, E. Stability of graph theoretical measures in structural brain networks in Alzheimer’s disease. Scientific Reports 2018, 8. [Google Scholar] [CrossRef] [PubMed]
  181. Shao, C.Y.; Mirra, S.S.; Sait, H.B.R.; Sacktor, T.C.; Sigurdsson, E.M. Postsynaptic degeneration as revealed by PSD-95 reduction occurs after advanced Aβ and tau pathology in transgenic mouse models of Alzheimer’s disease. Acta Neuropathologica 2011, 122, 285–292. [Google Scholar] [CrossRef] [PubMed]
  182. Savioz, A.; Leuba, G.; Vallet, P.G. A framework to understand the variations of PSD-95 expression in brain aging and in Alzheimer’s disease. Ageing Research Reviews 2014, 18, 86–94. [Google Scholar] [CrossRef] [PubMed]
  183. Kivisäkk, P.; Carlyle, B.C.; Sweeney, T.; Quinn, J.P.; Ramirez, C.E.; Trombetta, B.A.; Mendes, M.; Brock, M.; Rubel, C.; Czerkowicz, J.; Graham, D.; Arnold, S.E. Increased levels of the synaptic proteins PSD-95, SNAP-25, and neurogranin in the cerebrospinal fluid of patients with Alzheimer’s disease. Alzheimer’s Research and Therapy 2022, 14. [Google Scholar] [CrossRef] [PubMed]
  184. Self, W.K.; Holtzman, D.M. Emerging diagnostics and therapeutics for Alzheimer disease. Nature Medicine 2023, 29, 2187–2199. [Google Scholar] [CrossRef] [PubMed]
  185. Talarico, G.; Trebbastoni, A.; Bruno, G.; de Lena, C. Modulation of the Cannabinoid System: A New Perspective for the Treatment of the Alzheimer’s Disease. Current Neuropharmacology 2019, 17, 176–183. [Google Scholar] [CrossRef] [PubMed]
  186. Irastorza, I.T. Glial localization of the cannabinoid CB1 and CB2 receptors in a mouse model of Alzheimer’s disease. PhD thesis, Neuroscience Department, Faculty of Medicine and Nursery, Universidad del País Vasco / Euskal Herriko Unibersitatea (UPV/EHU), 2021.
  187. Kane, J.P.M.; Surendranathan, A.; Bentley, A.; Barker, S.A.H.; Taylor, J.P.; Thomas, A.J.; Allan, L.M.; McNally, R.J.; James, P.W.; McKeith, I.G.; Burn, D.J.; O’Brien, J.T. Clinical prevalence of Lewy body dementia. Alzheimer’s Research and Therapy 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  188. Fahn, S. Classification of movement disorders. Movement Disorders 2011, 26, 947–957. [Google Scholar] [CrossRef] [PubMed]
  189. Abdo, W.F.; van de Warrenburg, B.P.C.; Burn, D.J.; Quinn, N.P.; Bloem, B.R. The clinical approach to movement disorders. Nature Reviews Neurology 2010, 6, 29–37. [Google Scholar] [CrossRef] [PubMed]
  190. Heim, B.; Krismer, F.; Marzi, R.D.; Seppi, K. Magnetic resonance imaging for the diagnosis of Parkinson’s disease. Journal of Neural Transmission 2017, 124, 915–964. [Google Scholar] [CrossRef]
  191. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nature Reviews Disease Primers 2017, 3. [Google Scholar] [CrossRef]
  192. Pietracupa, S.; Martin-Bastida, A.; Piccini, P. Iron metabolism and its detection through MRI in parkinsonian disorders: a systematic review. Neurological Sciences 2017, 38, 2095–2101. [Google Scholar] [CrossRef]
  193. Dean, D.C.; Sojkova, J.; Hurley, S.; Kecskemeti, S.; Okonkwo, O.; Bendlin, B.B.; Theisen, F.; Johnson, S.C.; Alexander, A.L.; Gallagher, C.L. Alterations of Myelin Content in Parkinson’s Disease: A Cross-Sectional Neuroimaging Study. PLOS ONE 2016, 11, e0163774. [Google Scholar] [CrossRef] [PubMed]
  194. Boshkovski, T.; Cohen-Adad, J.; Misic, B.; Arnulf, I.; Corvol, J.C.; Vidailhet, M.; Lehéricy, S.; Stikov, N.; Mancini, M. The Myelin-Weighted Connectome in Parkinson’s Disease. Movement Disorders 2021, 37, 724–733. [Google Scholar] [CrossRef] [PubMed]
  195. Yang, Y.; Ye, C.; Sun, J.; Liang, L.; Lv, H.; Gao, L.; Fang, J.; Ma, T.; Wu, T. Alteration of brain structural connectivity in progression of Parkinson’s disease: A connectome-wide network analysis. NeuroImage: Clinical 2021, 31, 102715. [Google Scholar] [CrossRef]
  196. Tinaz, S. Functional Connectome in Parkinson’s Disease and Parkinsonism. Current Neurology and Neuroscience Reports 2021, 21. [Google Scholar] [CrossRef]
  197. Loh, A.; Boutet, A.; Germann, J.; Al-Fatly, B.; Elias, G.J.B.; Neudorfer, C.; Krotz, J.; Wong, E.H.Y.; Parmar, R.; Gramer, R.; Paff, M.; Horn, A.; Chen, J.J.; Azevedo, P.; Fasano, A.; Munhoz, R.P.; Hodaie, M.; Kalia, S.K.; Kucharczyk, W.; Lozano, A.M. A Functional Connectome of Parkinson’s Disease Patients Prior to Deep Brain Stimulation: A Tool for Disease-Specific Connectivity Analyses. Frontiers in Neuroscience 2022, 16. [Google Scholar] [CrossRef]
  198. Ashizawa, T.; Xia, G. Ataxia. CONTINUUM: Lifelong Learning in Neurology 2016, 22, 1208–1226. [Google Scholar] [CrossRef] [PubMed]
  199. Walker, F.O. Huntington’s disease. The Lancet 2007, 369, 218–228. [Google Scholar] [CrossRef]
  200. Roos, R.A. Huntington’s disease: a clinical review. Orphanet Journal of Rare Diseases 2010, 5. [Google Scholar] [CrossRef]
  201. Odish, O.F.; Caeyenberghs, K.; Hosseini, H.; van den Bogaard, S.J.; Roos, R.A.; Leemans, A. Dynamics of the connectome in Huntington’s disease: A longitudinal diffusion MRI study. NeuroImage: Clinical 2015, 9, 32–43. [Google Scholar] [CrossRef]
  202. Poudel, G.R.; Harding, I.H.; Egan, G.F.; Georgiou-Karistianis, N. Network spread determines severity of degeneration and disconnection in Huntington’s disease. Human Brain Mapping 2019, 40, 4192–4201. [Google Scholar] [CrossRef]
  203. Espinoza, F.A.; Turner, J.A.; Vergara, V.M.; Miller, R.L.; Mennigen, E.; Liu, J.; Misiura, M.B.; Ciarochi, J.; Johnson, H.J.; Long, J.D.; Bockholt, H.J.; Magnotta, V.A.; Paulsen, J.S.; Calhoun, V.D. Whole-Brain Connectivity in a Large Study of Huntington’s Disease Gene Mutation Carriers and Healthy Controls. Brain Connectivity 2018, 8, 166–178. [Google Scholar] [CrossRef] [PubMed]
  204. Scheckel, C.; Aguzzi, A. Prions, prionoids and protein misfolding disorders. Nature Reviews Genetics 2018, 19, 405–418. [Google Scholar] [CrossRef] [PubMed]
  205. Johnson, R.T. Prion diseases. The Lancet Neurology 2005, 4, 635–642. [Google Scholar] [CrossRef] [PubMed]
  206. Knight, R.S.G. PRION DISEASES. Journal of Neurology, Neurosurgery and Psychiatry 2004, 75, 36i–342. [Google Scholar] [CrossRef] [PubMed]
  207. Ironside, J.W.; Ritchie, D.L.; Head, M.W. , Prion diseases. In Neuropathology; Elsevier, 2018; p. 393–403. [CrossRef]
  208. Fornari, S.; Schäfer, A.; Jucker, M.; Goriely, A.; Kuhl, E. Prion-like spreading of Alzheimer’s disease within the brain’s connectome. Journal of The Royal Society Interface 2019, 16, 20190356. [Google Scholar] [CrossRef] [PubMed]
  209. Chaudhuri, A. Multiple sclerosis is primarily a neurodegenerative disease. Journal of Neural Transmission 2013, 120, 1463–1466. [Google Scholar] [CrossRef] [PubMed]
  210. Gironi, M.; Arnò, C.; Comi, G.; Penton-Rol, G.; Furlan, R. , Multiple Sclerosis and Neurodegenerative Diseases. In Immune Rebalancing; Elsevier, 2016; p. 63–84. [CrossRef]
  211. Goldenberg, M.M. Multiple Sclerosis Review. Pharmacy and Therapeutics 2012. [Google Scholar]
  212. Dobson, R.; Giovannoni, G. Multiple sclerosis – a review. European Journal of Neurology 2018, 26, 27–40. [Google Scholar] [CrossRef]
  213. Schoonheim, M.M.; Meijer, K.A.; Geurts, J.J.G. Network Collapse and Cognitive Impairment in Multiple Sclerosis. Frontiers in Neurology 2015, 6. [Google Scholar] [CrossRef]
  214. Høgestøl, E.A.; Ghezzo, S.; Nygaard, G.O.; Espeseth, T.; Sowa, P.; Beyer, M.K.; Harbo, H.F.; Westlye, L.T.; Hulst, H.E.; Alnæs, D. Functional connectivity in multiple sclerosis modelled as connectome stability: A 5-year follow-up study. Multiple Sclerosis Journal 2021, 28, 532–540. [Google Scholar] [CrossRef]
  215. Schoonheim, M.M.; Broeders, T.A.; Geurts, J.J. The network collapse in multiple sclerosis: An overview of novel concepts to address disease dynamics. NeuroImage: Clinical 2022, 35, 103108. [Google Scholar] [CrossRef]
  216. Barile, B.; Ashtari, P.; Stamile, C.; Marzullo, A.; Maes, F.; Durand-Dubief, F.; Van Huffel, S.; Sappey-Marinier, D. Classification of multiple sclerosis clinical profiles using machine learning and grey matter connectome. Frontiers in Robotics and AI 2022, 9. [Google Scholar] [CrossRef]
  217. Manglani, H.R.; Fountain-Zaragoza, S.; Shankar, A.; Nicholas, J.A.; Prakash, R.S. Employing connectome-based models to predict working memory in multiple sclerosis. Brain Connectivity 2021, 12. [Google Scholar] [CrossRef]
  218. Tahedl, M.; Levine, S.M.; Greenlee, M.W.; Weissert, R.; Schwarzbach, J.V. Functional Connectivity in Multiple Sclerosis: Recent Findings and Future Directions. Frontiers in Neurology 2018, 9. [Google Scholar] [CrossRef]
  219. Hauser, S.L.; Cree, B.A. Treatment of Multiple Sclerosis: A Review. The American Journal of Medicine 2020, 133, 1380–1390. [Google Scholar] [CrossRef]
  220. Feldman, E.L.; Goutman, S.A.; Petri, S.; Mazzini, L.; Savelieff, M.G.; Shaw, P.J.; Sobue, G. Amyotrophic lateral sclerosis. The Lancet 2022, 400, 1363–1380. [Google Scholar] [CrossRef]
  221. Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nature Reviews Disease Primers 2017, 3. [Google Scholar] [CrossRef] [PubMed]
  222. Masrori, P.; Van Damme, P. Amyotrophic lateral sclerosis: a clinical review. European Journal of Neurology 2020, 27, 1918–1929. [Google Scholar] [CrossRef] [PubMed]
  223. Sorrentino, P.; Rucco, R.; Jacini, F.; Trojsi, F.; Lardone, A.; Baselice, F.; Femiano, C.; Santangelo, G.; Granata, C.; Vettoliere, A.; Monsurrò, M.R.; Tedeschi, G.; Sorrentino, G. Brain functional networks become more connected as amyotrophic lateral sclerosis progresses: a source level magnetoencephalographic study. NeuroImage: Clinical 2018, 20, 564–571. [Google Scholar] [CrossRef]
  224. Schmidt, R.; Verstraete, E.; de Reus, M.A.; Veldink, J.H.; van den Berg, L.H.; van den Heuvel, M.P. Correlation between structural and functional connectivity impairment in amyotrophic lateral sclerosis. Human Brain Mapping 2014, 35, 4386–4395. [Google Scholar] [CrossRef]
  225. Verstraete, E.; van den Heuvel, M.P.; Veldink, J.H.; Blanken, N.; Mandl, R.C.; Hulshoff Pol, H.E.; van den Berg, L.H. Motor Network Degeneration in Amyotrophic Lateral Sclerosis: A Structural and Functional Connectivity Study. PLoS ONE 2010, 5, e13664. [Google Scholar] [CrossRef]
  226. Agosta, F.; Canu, E.; Valsasina, P.; Riva, N.; Prelle, A.; Comi, G.; Filippi, M. Divergent brain network connectivity in amyotrophic lateral sclerosis. Neurobiology of Aging 2013, 34, 419–427. [Google Scholar] [CrossRef]
  227. Kessler, R.C.; Amminger, G.P.; Aguilar-Gaxiola, S.; Alonso, J.; Lee, S.; ??st??n, T.B. Age of onset of mental disorders: a review of recent literature. Current Opinion in Psychiatry 2007, 20, 359–364. [Google Scholar] [CrossRef]
  228. Solmi, M.; Radua, J.; Olivola, M.; Croce, E.; Soardo, L.; Salazar de Pablo, G.; Il Shin, J.; Kirkbride, J.B.; Jones, P.; Kim, J.H.; Kim, J.Y.; Carvalho, A.F.; Seeman, M.V.; Correll, C.U.; Fusar-Poli, P. Age at onset of mental disorders worldwide: large-scale meta-analysis of 192 epidemiological studies. Molecular Psychiatry 2021, 27, 281–295. [Google Scholar] [CrossRef]
  229. Crossley, N.A.; Mechelli, A.; Vértes, P.E.; Winton-Brown, T.T.; Patel, A.X.; Ginestet, C.E.; McGuire, P.; Bullmore, E.T. Cognitive relevance of the community structure of the human brain functional coactivation network. Proceedings of the National Academy of Sciences 2013, 110, 11583–11588. [Google Scholar] [CrossRef]
  230. Crossley, N.A. , Connectome analysis and psychiatric disorders. In Connectome Analysis; Elsevier, 2023; p. 423–432. [CrossRef]
  231. Van Essen, D.C.; Barch, D.M. The human connectome in health and psychopathology. World Psychiatry 2015, 14, 154–157. [Google Scholar] [CrossRef]
  232. van den Heuvel, M.P.; Sporns, O. A cross-disorder connectome landscape of brain dysconnectivity. Nature Reviews Neuroscience 2019, 20, 435–446. [Google Scholar] [CrossRef] [PubMed]
  233. Solmi, M.; Seitidis, G.; Mavridis, D.; Correll, C.U.; Dragioti, E.; Guimond, S.; Tuominen, L.; Dargél, A.; Carvalho, A.F.; Fornaro, M.; Maes, M.; Monaco, F.; Song, M.; Il Shin, J.; Cortese, S. Incidence, prevalence, and global burden of schizophrenia - data, with critical appraisal, from the Global Burden of Disease (GBD) 2019. Molecular Psychiatry 2023. [Google Scholar] [CrossRef] [PubMed]
  234. Schultz, S.H.; North, S.W.; Shields, C.G. Schizophrenia: A Review. American Family Physician 2007, 75, 1821–1829. [Google Scholar] [PubMed]
  235. Patel, K.R.; Cherian, J.; Gohil, K.; Atkinson, D. Schizophrenia: Overview and Treatment Options. Pharmacy and Therapeutics 2014. [Google Scholar]
  236. Yuan, L.; Ma, X.; Li, D.; Ouyang, L.; Fan, L.; Li, C.; He, Y.; Chen, X. Alteration of a brain network with stable and strong functional connections in subjects with schizophrenia. Schizophrenia 2022, 8. [Google Scholar] [CrossRef] [PubMed]
  237. Rubinov, M.; Knock, S.A.; Stam, C.J.; Micheloyannis, S.; Harris, A.W.; Williams, L.M.; Breakspear, M. Small-world properties of nonlinear brain activity in schizophrenia. Human Brain Mapping 2009, 30, 403–416. [Google Scholar] [CrossRef] [PubMed]
  238. Micheloyannis, S.; Pachou, E.; Stam, C.J.; Breakspear, M.; Bitsios, P.; Vourkas, M.; Erimaki, S.; Zervakis, M. Small-world networks and disturbed functional connectivity in schizophrenia. Schizophrenia Research 2006, 87, 60–66. [Google Scholar] [CrossRef] [PubMed]
  239. Mastrandrea, R.; Piras, F.; Gabrielli, A.; Banaj, N.; Caldarelli, G.; Spalletta, G.; Gili, T. The unbalanced reorganization of weaker functional connections induces the altered brain network topology in schizophrenia. Scientific Reports 2021, 11. [Google Scholar] [CrossRef]
  240. Belmaker, R.; Agam, G. Major Depressive Disorder. New England Journal of Medicine 2008, 358, 55–68. [Google Scholar] [CrossRef]
  241. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major depressive disorder. Nature Reviews Disease Primers 2016, 2. [Google Scholar] [CrossRef] [PubMed]
  242. Scheuer, H.; Alarcón, G.; Demeter, D.V.; Earl, E.; Fair, D.A.; Nagel, B.J. Reduced fronto-amygdalar connectivity in adolescence is associated with increased depression symptoms over time. Psychiatry Research: Neuroimaging 2017, 266, 35–41. [Google Scholar] [CrossRef]
  243. Liu, J.; Fan, Y.; Zeng, L.L.; Liu, B.; Ju, Y.; Wang, M.; Dong, Q.; Lu, X.; Sun, J.; Zhang, L.; Guo, H.; Zhao, F.; Li, W.; Zhang, L.; Li, Z.; Liao, M.; Zhang, Y.; Hu, D.; Li, L. The neuroprogressive nature of major depressive disorder: evidence from an intrinsic connectome analysis. Translational Psychiatry 2021, 11. [Google Scholar] [CrossRef]
  244. Yang, H.; Chen, X.; Chen, Z.B.; Li, L.; Li, X.Y.; Castellanos, F.X.; Bai, T.J.; Bo, Q.J.; Cao, J.; Chang, Z.K.; Chen, G.M.; Chen, N.X.; Chen, W.; Cheng, C.; Cheng, Y.Q.; Cui, X.L.; Duan, J.; Fang, Y.; Gong, Q.Y.; Guo, W.B.; Hou, Z.H.; Hu, L.; Kuang, L.; Li, F.; Li, H.X.; Li, K.M.; Li, T.; Liu, Y.S.; Liu, Z.N.; Long, Y.C.; Lu, B.; Luo, Q.H.; Meng, H.Q.; Peng, D.; Qiu, H.T.; Qiu, J.; Shen, Y.D.; Shi, Y.S.; Si, T.M.; Tang, Y.Q.; Wang, C.Y.; Wang, F.; Wang, K.; Wang, L.; Wang, X.; Wang, Y.; Wang, Y.W.; Wu, X.P.; Wu, X.R.; Xie, C.M.; Xie, G.R.; Xie, H.Y.; Xie, P.; Xu, X.F.; Yang, J.; Yao, J.S.; Yao, S.Q.; Yin, Y.Y.; Yuan, Y.G.; Zang, Y.F.; Zhang, A.X.; Zhang, H.; Zhang, K.R.; Zhang, L.; Zhang, Z.J.; Zhao, J.P.; Zhou, R.; Zhou, Y.T.; Zhu, J.J.; Zhu, Z.C.; Zou, C.J.; Zuo, X.N.; Yan, C.G. Disrupted intrinsic functional brain topology in patients with major depressive disorder. Molecular Psychiatry 2021, 26, 7363–7371. [Google Scholar] [CrossRef] [PubMed]
  245. Sikora, M.; Heffernan, J.; Avery, E.T.; Mickey, B.J.; Zubieta, J.K.; Peciña, M. Salience Network Functional Connectivity Predicts Placebo Effects in Major Depression. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging 2016, 1, 68–76. [Google Scholar] [CrossRef] [PubMed]
  246. Scalabrini, A.; Vai, B.; Poletti, S.; Damiani, S.; Mucci, C.; Colombo, C.; Zanardi, R.; Benedetti, F.; Northoff, G. All roads lead to the default-mode network—global source of DMN abnormalities in major depressive disorder. Neuropsychopharmacology 2020, 45, 2058–2069. [Google Scholar] [CrossRef] [PubMed]
  247. Saris, I.M.J.; Penninx, B.W.J.H.; Dinga, R.; van Tol, M.J.; Veltman, D.J.; van der Wee, N.J.A.; Aghajani, M. Default Mode Network Connectivity and Social Dysfunction in Major Depressive Disorder. Scientific Reports 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  248. Hill, A.T.; Hadas, I.; Zomorrodi, R.; Voineskos, D.; Farzan, F.; Fitzgerald, P.B.; Blumberger, D.M.; Daskalakis, Z.J. Modulation of functional network properties in major depressive disorder following electroconvulsive therapy (ECT): a resting-state EEG analysis. Scientific Reports 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  249. Korgaonkar, M.S.; Goldstein-Piekarski, A.N.; Fornito, A.; Williams, L.M. Intrinsic connectomes are a predictive biomarker of remission in major depressive disorder. Molecular Psychiatry 2019, 25, 1537–1549. [Google Scholar] [CrossRef] [PubMed]
  250. Berwian, I.M.; Wenzel, J.G.; Kuehn, L.; Schnuerer, I.; Kasper, L.; Veer, I.M.; Seifritz, E.; Stephan, K.E.; Walter, H.; Huys, Q.J.M. The relationship between resting-state functional connectivity, antidepressant discontinuation and depression relapse. Scientific Reports 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  251. Repple, J.; Mauritz, M.; Meinert, S.; de Lange, S.C.; Grotegerd, D.; Opel, N.; Redlich, R.; Hahn, T.; Förster, K.; Leehr, E.J.; Winter, N.; Goltermann, J.; Enneking, V.; Fingas, S.M.; Lemke, H.; Waltemate, L.; Nenadic, I.; Krug, A.; Brosch, K.; Schmitt, S.; Stein, F.; Meller, T.; Jansen, A.; Steinsträter, O.; Baune, B.T.; Kircher, T.; Dannlowski, U.; van den Heuvel, M.P. Severity of current depression and remission status are associated with structural connectome alterations in major depressive disorder. Molecular Psychiatry 2019, 25, 1550–1558. [Google Scholar] [CrossRef] [PubMed]
  252. Fava, M.; Kendler, K.S. Major Depressive Disorder. Neuron 2000, 28, 335–341. [Google Scholar] [CrossRef]
  253. Carvalho, A.F.; Firth, J.; Vieta, E. Bipolar Disorder. New England Journal of Medicine 2020, 383, 58–66. [Google Scholar] [CrossRef]
  254. Vieta, E.; Berk, M.; Schulze, T.G.; Carvalho, A.F.; Suppes, T.; Calabrese, J.R.; Gao, K.; Miskowiak, K.W.; Grande, I. Bipolar disorders. Nature Reviews Disease Primers 2018, 4. [Google Scholar] [CrossRef] [PubMed]
  255. Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar disorder. The Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef]
  256. Jiang, H.; Zhu, R.; Tian, S.; Wang, H.; Chen, Z.; Wang, X.; Shao, J.; Qin, J.; Shi, J.; Liu, H.; Chen, Y.; Yao, Z.; Lu, Q. Structural–functional decoupling predicts suicide attempts in bipolar disorder patients with a current major depressive episode. Neuropsychopharmacology 2020, 45, 1735–1742. [Google Scholar] [CrossRef]
  257. Sankar, A.; Scheinost, D.; Goldman, D.A.; Drachman, R.; Colic, L.; Villa, L.M.; Kim, J.A.; Gonzalez, Y.; Marcelo, I.; Shinomiya, M.; Pittman, B.; Lacadie, C.M.; Oquendo, M.A.; Constable, R.T.; Blumberg, H.P. Graph theory analysis of whole brain functional connectivity to assess disturbances associated with suicide attempts in bipolar disorder. Translational Psychiatry 2022, 12. [Google Scholar] [CrossRef] [PubMed]
  258. Fombonne, E. Epidemiology of Autistic Disorder and Other Pervasive Developmental Disorders. The Journal of Clinical Psychiatry 2005. [Google Scholar]
  259. Lord, C.; Brugha, T.S.; Charman, T.; Cusack, J.; Dumas, G.; Frazier, T.; Jones, E.J.H.; Jones, R.M.; Pickles, A.; State, M.W.; Taylor, J.L.; Veenstra-VanderWeele, J. Autism spectrum disorder. Nature Reviews Disease Primers 2020, 6. [Google Scholar] [CrossRef] [PubMed]
  260. Rutter, M. CONCEPTS OF AUTISM: A REVIEW OF RESEARCH*. Journal of Child Psychology and Psychiatry 1968, 9, 1–25. [Google Scholar] [CrossRef]
  261. Zeidan, J.; Fombonne, E.; Scorah, J.; Ibrahim, A.; Durkin, M.S.; Saxena, S.; Yusuf, A.; Shih, A.; Elsabbagh, M. Global prevalence of autism: A systematic review update. Autism Research 2022, 15, 778–790. [Google Scholar] [CrossRef]
  262. FOMBONNE, E. The epidemiology of autism: a review. Psychological Medicine 1999, 29, 769–786. [Google Scholar] [CrossRef]
  263. Miles, J.H. Autism spectrum disorders—A genetics review. Genetics in Medicine 2011, 13, 278–294. [Google Scholar] [CrossRef]
  264. Mohammad-Rezazadeh, I.; Frohlich, J.; Loo, S.K.; Jeste, S.S. Brain connectivity in autism spectrum disorder. Current Opinion in Neurology 2016, 29, 137–147. [Google Scholar] [CrossRef] [PubMed]
  265. Berto, S.; Treacher, A.H.; Caglayan, E.; Luo, D.; Haney, J.R.; Gandal, M.J.; Geschwind, D.H.; Montillo, A.A.; Konopka, G. Association between resting-state functional brain connectivity and gene expression is altered in autism spectrum disorder. Nature Communications 2022. [Google Scholar] [CrossRef] [PubMed]
  266. Kana, R.K.; Uddin, L.Q.; Kenet, T.; Chigani, D.; Müller, R.A. Brain connectivity in autism. Frontiers in Human Neuroscience 2014. [Google Scholar] [CrossRef] [PubMed]
  267. Benkarim, O.; Paquola, C.; Park, B.y.; Hong, S.J.; Royer, J.; Vos de Wael, R.; Lariviere, S.; Valk, S.; Bzdok, D.; Mottron, L.; C. Bernhardt, B. Connectivity alterations in autism reflect functional idiosyncrasy. Communications Biology 2021, 4. [Google Scholar] [CrossRef]
  268. Liu, X.; Huang, H. Alterations of functional connectivities associated with autism spectrum disorder symptom severity: a multi-site study using multivariate pattern analysis. Scientific Reports 2020, 10. [Google Scholar] [CrossRef]
  269. Redcay, E.; Moran, J.M.; Mavros, P.L.; Tager-Flusberg, H.; Gabrieli, J.D.E.; Whitfield-Gabrieli, S. Intrinsic functional network organization in high-functioning adolescents with autism spectrum disorder. Frontiers in Human Neuroscience 2013, 7. [Google Scholar] [CrossRef] [PubMed]
  270. Roine, U.; Roine, T.; Salmi, J.; Nieminen-von Wendt, T.; Tani, P.; Leppämäki, S.; Rintahaka, P.; Caeyenberghs, K.; Leemans, A.; Sams, M. Abnormal wiring of the connectome in adults with high-functioning autism spectrum disorder. Molecular Autism 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  271. Vissers, M.E.; X Cohen, M.; Geurts, H.M. Brain connectivity and high functioning autism: A promising path of research that needs refined models, methodological convergence, and stronger behavioral links. Neuroscience and Biobehavioral Reviews 2012, 36, 604–625. [Google Scholar] [CrossRef] [PubMed]
  272. Woodbury-Smith, M.R.; Volkmar, F.R. Asperger syndrome. European Child and Adolescent Psychiatry 2008, 18, 2–11. [Google Scholar] [CrossRef]
  273. Tantam, D. Asperger’S Syndrome. Journal of Child Psychology and Psychiatry 1988, 29, 245–255. [Google Scholar] [CrossRef]
  274. Tantam, D. Psychological Disorder in Adolescents and Adults with Asperger Syndrome. Autism 2000, 4, 47–62. [Google Scholar] [CrossRef]
  275. Javaheripour, N.; Wagner, G.; de la Cruz, F.; Walter, M.; Szycik, G.R.; Tietze, F.A. Altered brain network organization in adults with Asperger’s syndrome: decreased connectome transitivity and assortativity with increased global efficiency. Frontiers in Psychiatry 2023, 14. [Google Scholar] [CrossRef] [PubMed]
  276. Stein, D.J.; Costa, D.L.C.; Lochner, C.; Miguel, E.C.; Reddy, Y.C.J.; Shavitt, R.G.; van den Heuvel, O.A.; Simpson, H.B. Obsessive–compulsive disorder. Nature Reviews Disease Primers 2019, 5. [Google Scholar] [CrossRef] [PubMed]
  277. Jenike, M.A. Obsessive–Compulsive Disorder. New England Journal of Medicine 2004, 350, 259–265. [Google Scholar] [CrossRef] [PubMed]
  278. Vogel, J.W.; Corriveau-Lecavalier, N.; Franzmeier, N.; Pereira, J.B.; Brown, J.A.; Maass, A.; Botha, H.; Seeley, W.W.; Bassett, D.S.; Jones, D.T.; Ewers, M. Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight. Nature Reviews Neuroscience 2023, 24, 620–639. [Google Scholar] [CrossRef]
  279. Chinesta, F.; Cueto, E.; Abisset-Chavanne, E.; Duval, J.L.; Khaldi, F.E. Virtual, Digital and Hybrid Twins: A New Paradigm in Data-Based Engineering and Engineered Data. Archives of Computational Methods in Engineering 2018, 27, 105–134. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated